Junqi Cui, Xia Wang, Jiali Lin, Yun Zou, Hongkang Wang, Wenyan Jiang, Jiajia Chen, Yamin Rao, Bin Xu, Long Li
Cystoscopy is the current gold standard for diagnosing bladder cancer (Bca), but its invasive nature often results in patient discomfort. This study aims to debise a new strategy to enhance BCa detection. We collected data from 30 BCa patients between January and June 2022. We obtained spontaneously voided urine specimens from these patients before and after administering extracorporeal bladder vibration. These specimens underwent routine cytologic examination and fluorescence in situ hybridization (FISH). Furthermore, we conducted a follow-up 3 months postoperation. We recollected urine specimens before and after extracorporeal bladder vibration, repeating the cytologic examination and FISH. Our findings indicated an increase in the number of exfoliated cells in patients' urine postvibration compared to previbration. The liquid-based cell staining results showed an increased detection rate of abnormal cells in the urinary sediment of patients with both low and high-grade urothelial carcinoma postvibration. Similarly, the FISH results revealed an increased detection rate of CEP3 and CEP7 positive cells postvibration. Additionally, 3 months postoperation, we found abnormal cells in the urine of one patient previbration and in three patients postvibration. Further cystoscopic biopsy confirmed that these three patients had developed tumor recurrence. Our study substantiates that the extracorporeal bladder vibration assay significantly enhances BCa detection and the prediction of tumor recurrence. This method is simple, quick, and cost-effective, making it a promising approach worthy of widespread clinical application.
{"title":"Extracorporeal bladder vibration assay enables early detection and recurrence monitoring for bladder cancer","authors":"Junqi Cui, Xia Wang, Jiali Lin, Yun Zou, Hongkang Wang, Wenyan Jiang, Jiajia Chen, Yamin Rao, Bin Xu, Long Li","doi":"10.1002/mog2.45","DOIUrl":"https://doi.org/10.1002/mog2.45","url":null,"abstract":"<p>Cystoscopy is the current gold standard for diagnosing bladder cancer (Bca), but its invasive nature often results in patient discomfort. This study aims to debise a new strategy to enhance BCa detection. We collected data from 30 BCa patients between January and June 2022. We obtained spontaneously voided urine specimens from these patients before and after administering extracorporeal bladder vibration. These specimens underwent routine cytologic examination and fluorescence in situ hybridization (FISH). Furthermore, we conducted a follow-up 3 months postoperation. We recollected urine specimens before and after extracorporeal bladder vibration, repeating the cytologic examination and FISH. Our findings indicated an increase in the number of exfoliated cells in patients' urine postvibration compared to previbration. The liquid-based cell staining results showed an increased detection rate of abnormal cells in the urinary sediment of patients with both low and high-grade urothelial carcinoma postvibration. Similarly, the FISH results revealed an increased detection rate of CEP3 and CEP7 positive cells postvibration. Additionally, 3 months postoperation, we found abnormal cells in the urine of one patient previbration and in three patients postvibration. Further cystoscopic biopsy confirmed that these three patients had developed tumor recurrence. Our study substantiates that the extracorporeal bladder vibration assay significantly enhances BCa detection and the prediction of tumor recurrence. This method is simple, quick, and cost-effective, making it a promising approach worthy of widespread clinical application.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 3","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.45","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50153629","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pu Luo, Jingjing Duan, Qiong Chen, Ling Shao, Wen Xiao, Xunqi Liu, Gengwei Zhang, Xiaohua Tan, Zhongyi Fan
Uveal melanoma (UM) is the most common primary intraocular malignant tumor in adults. More than half of UM cases develop distant metastasis to the liver, lung, bone, and other organs, which frequently results in patient death. However, the mechanisms underlying UM metastasis remain largely unknown. Here, we report that protein tyrosine kinase 2 (PTK2), a nonreceptor protein tyrosine kinase also known as focal adhesion kinase (FAK), was overexpressed in most examined UM specimens. Furthermore, we identified PTK2 expression as a novel independent risk factor that predicts poor prognosis of UM patients. Mechanistic studies revealed that PTK2 promotes the EMT phenotype, leading to metastasis of UM cells. We showed that PTK2, located on chromosome 8q, is a functional gene of chromosome 8q gain to UM metastasis, which may represent the molecular mechanism for the aberrant expression and activation of PTK2 in UM. Our data reveal a novel role and mechanism of PTK2 in the metastatic process of UM, suggesting PTK2 may be a potential prognostic biomarker for UM metastasis and a promising therapeutic target for UM treatment.
{"title":"PTK2 promotes uveal melanoma metastasis by activating epithelial-to-mesenchymal transition","authors":"Pu Luo, Jingjing Duan, Qiong Chen, Ling Shao, Wen Xiao, Xunqi Liu, Gengwei Zhang, Xiaohua Tan, Zhongyi Fan","doi":"10.1002/mog2.44","DOIUrl":"https://doi.org/10.1002/mog2.44","url":null,"abstract":"<p>Uveal melanoma (UM) is the most common primary intraocular malignant tumor in adults. More than half of UM cases develop distant metastasis to the liver, lung, bone, and other organs, which frequently results in patient death. However, the mechanisms underlying UM metastasis remain largely unknown. Here, we report that protein tyrosine kinase 2 (PTK2), a nonreceptor protein tyrosine kinase also known as focal adhesion kinase (FAK), was overexpressed in most examined UM specimens. Furthermore, we identified PTK2 expression as a novel independent risk factor that predicts poor prognosis of UM patients. Mechanistic studies revealed that PTK2 promotes the EMT phenotype, leading to metastasis of UM cells. We showed that PTK2, located on chromosome 8q, is a functional gene of chromosome 8q gain to UM metastasis, which may represent the molecular mechanism for the aberrant expression and activation of PTK2 in UM. Our data reveal a novel role and mechanism of PTK2 in the metastatic process of UM, suggesting PTK2 may be a potential prognostic biomarker for UM metastasis and a promising therapeutic target for UM treatment.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 3","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.44","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50136893","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fan Liao, Rui-Xi Hua, Xingyu Jia, Yuxiang Liao, Li Yuan, Jichen Ruan, Tianfeng Li, Zhenjian Zhuo, Jing He
Glioma is a highly heterogeneous malignancy with a high mortality rate and poor prognosis. m1A methylation modifications are associated with gliomagenesis. However, whether single nucleotide polymorphisms (SNPs) of m1A modification genes are associated with glioma risk is unclear. We successfully genotyped 20 SNPs of m1A-modified genes TRMT10C, TRMT61B, TRMT6, TRM61, ALKBH1, YTHDC1, YTHDF1, and YTHDF2 in 314 pediatric glioma patients and 380 cancer-free controls using TaqMan probes. Associations of polymorphisms with glioma risk were assessed by the odds ratios and 95% confidence intervals generated by logistic regression models. Stratified analysis was performed by age, gender, tumor subtype, and clinical stage. The results showed that TRMT10C rs2303476, TRMT10C rs4257518, TRM61 rs2296484, and YTHDF2 rs3738067 polymorphisms were significantly associated with an increased risk of glioma, TRMT61B rs4563180, YTHDC1 rs2293595, and YTHDC1 rs3813832 polymorphisms were significantly associated with a reduced risk of glioma. In addition, analysis of the expression quantitative trait loci-showed that the TRM61 rs2296484 T allele significantly increased TRM61 messenger RNA (mRNA) expression, the YTHDF2 rs3738067 G allele significantly increased YTHDF2 mRNA expression, and the TRMT61B rs4563180 C allele significantly decreased TRMT61B mRNA expression. Overall, we identified several promising candidates for m1A modification gene polymorphisms as biomarkers of glioma risk.
{"title":"Association of m1A modification gene polymorphisms with glioma risk in Chinese children","authors":"Fan Liao, Rui-Xi Hua, Xingyu Jia, Yuxiang Liao, Li Yuan, Jichen Ruan, Tianfeng Li, Zhenjian Zhuo, Jing He","doi":"10.1002/mog2.43","DOIUrl":"https://doi.org/10.1002/mog2.43","url":null,"abstract":"<p>Glioma is a highly heterogeneous malignancy with a high mortality rate and poor prognosis. m<sup>1</sup>A methylation modifications are associated with gliomagenesis. However, whether single nucleotide polymorphisms (SNPs) of m<sup>1</sup>A modification genes are associated with glioma risk is unclear. We successfully genotyped 20 SNPs of m<sup>1</sup>A-modified genes <i>TRMT10C, TRMT61B, TRMT6, TRM61, ALKBH1, YTHDC1, YTHDF1</i>, and <i>YTHDF2</i> in 314 pediatric glioma patients and 380 cancer-free controls using TaqMan probes. Associations of polymorphisms with glioma risk were assessed by the odds ratios and 95% confidence intervals generated by logistic regression models. Stratified analysis was performed by age, gender, tumor subtype, and clinical stage. The results showed that <i>TRMT10C</i> rs2303476, <i>TRMT10C</i> rs4257518, <i>TRM61</i> rs2296484, and <i>YTHDF2</i> rs3738067 polymorphisms were significantly associated with an increased risk of glioma, <i>TRMT61B</i> rs4563180, <i>YTHDC1</i> rs2293595, and <i>YTHDC1</i> rs3813832 polymorphisms were significantly associated with a reduced risk of glioma. In addition, analysis of the expression quantitative trait loci-showed that the <i>TRM61</i> rs2296484 T allele significantly increased <i>TRM61</i> messenger RNA (mRNA) expression, the <i>YTHDF2</i> rs3738067 G allele significantly increased <i>YTHDF2</i> mRNA expression, and the <i>TRMT61B</i> rs4563180 C allele significantly decreased <i>TRMT61B</i> mRNA expression. Overall, we identified several promising candidates for m<sup>1</sup>A modification gene polymorphisms as biomarkers of glioma risk.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 3","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.43","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50134566","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cuproptosis is a novel form of copper-induced programmed cell death. Here, we investigate the role of cuproptosis-related genes in head and neck squamous cell carcinoma (HNSCC), and the correlations between cuproptosis-related genes and tumor-infiltrating immune cells in the microenvironment. Gene expression data were downloaded from TCGA and analyzed using R. Protein–protein interaction analysis was conducted using STRING and GeneMANIA, while survival analyses examined the correlations between genes and overall survival. In addition, analyses of methylation and mutation sites were performed using Illumina HumanMethylation450 data and the cBioPortal data set, respectively. The correlations between gene expression level and infiltrating immune cells and predictive values of PD-1/PD-L1 blockade responses were also analyzed. Cuproptosis-promoting genes were downregulated, while two out of three cuproptosis-inhibiting genes, GLS and CDKN2A, showed upregulated expression. The CDKN2A expression level was identified as an independent prognostic factor for HNSCC through the multivariate Cox survival analysis. Meanwhile, levels of all cuproptosis-related genes correlated positively with CD4+ T cell infiltration. Furthermore, expression of the cuproptosis-promoting gene DLD was negatively correlated with immune score. All cuproptosis-related genes were expressed aberrantly and correlated positively with CD4+ T cell infiltration in HNSCC. Thus, copper homeostasis and cuproptosis could be targeted therapeutically as a potential means of enhancing the efficacy of immunotherapy.
{"title":"Comprehensive analysis of cuproptosis-related gene expression and positive correlations with CD4+ T cell infiltration in head and neck squamous cell carcinoma","authors":"Zhangwei Hu, Wenbin Lei, Weiping Wen","doi":"10.1002/mog2.42","DOIUrl":"https://doi.org/10.1002/mog2.42","url":null,"abstract":"<p>Cuproptosis is a novel form of copper-induced programmed cell death. Here, we investigate the role of cuproptosis-related genes in head and neck squamous cell carcinoma (HNSCC), and the correlations between cuproptosis-related genes and tumor-infiltrating immune cells in the microenvironment. Gene expression data were downloaded from TCGA and analyzed using R. Protein–protein interaction analysis was conducted using STRING and GeneMANIA, while survival analyses examined the correlations between genes and overall survival. In addition, analyses of methylation and mutation sites were performed using Illumina HumanMethylation450 data and the cBioPortal data set, respectively. The correlations between gene expression level and infiltrating immune cells and predictive values of PD-1/PD-L1 blockade responses were also analyzed. Cuproptosis-promoting genes were downregulated, while two out of three cuproptosis-inhibiting genes, GLS and CDKN2A, showed upregulated expression. The CDKN2A expression level was identified as an independent prognostic factor for HNSCC through the multivariate Cox survival analysis. Meanwhile, levels of all cuproptosis-related genes correlated positively with CD4<sup>+</sup> T cell infiltration. Furthermore, expression of the cuproptosis-promoting gene DLD was negatively correlated with immune score. All cuproptosis-related genes were expressed aberrantly and correlated positively with CD4<sup>+</sup> T cell infiltration in HNSCC. Thus, copper homeostasis and cuproptosis could be targeted therapeutically as a potential means of enhancing the efficacy of immunotherapy.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 3","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-07-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.42","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50123771","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Baiyang Liu, Yulin Shi, Bo Qin, Dating Cheng, Zhi Nie, Haoqing Zhai, Yao Li, Zhixian Jin, Jun Pu, Dongming Yan, Yongbin Chen, Cuiping Yang
Gliomas, the most lethal brain tumors, often exhibit resistance to conventional chemotherapy and/or radiotherapy. This study reveals that sertindole, a potent dopamine D2 receptor antagonist primarily designed as an antipsychotic medication for schizophrenia, effectively inhibits glioma progression. Our findings demonstrate that sertindole suppresses the proliferation of U251 and U87 tumor cells, impedes cell cycle progression in vitro, and curtails xenograft tumor growth in vivo. Moreover, we present compelling evidence demonstrating the ability of sertindole to enhance the cellular response to the chemotherapeutic agent temozolomide both in vitro and in vivo. Additionally, our findings reveal that sertindole effectively suppresses the self-renewal capacity and expression of stemness-associated genes, such as Nanog and Sox2, in glioma tumor cells and glioma stem cells. A mechanistic investigation demonstrated that sertindole enhances the formation of autophagosome–lysosome complexes while concurrently impeding autophagic flux through the inhibition of lysosomal hydrolytic enzymes CTSD and CTSB, ultimately resulting in decreased growth of tumor cells. In conclusion, our findings suggest that sertindole has the potential to develop into a potent antiglioma therapeutic agent.
{"title":"Sertindole inhibits autophagic flux and glioma progression","authors":"Baiyang Liu, Yulin Shi, Bo Qin, Dating Cheng, Zhi Nie, Haoqing Zhai, Yao Li, Zhixian Jin, Jun Pu, Dongming Yan, Yongbin Chen, Cuiping Yang","doi":"10.1002/mog2.41","DOIUrl":"https://doi.org/10.1002/mog2.41","url":null,"abstract":"<p>Gliomas, the most lethal brain tumors, often exhibit resistance to conventional chemotherapy and/or radiotherapy. This study reveals that sertindole, a potent dopamine D2 receptor antagonist primarily designed as an antipsychotic medication for schizophrenia, effectively inhibits glioma progression. Our findings demonstrate that sertindole suppresses the proliferation of U251 and U87 tumor cells, impedes cell cycle progression in vitro, and curtails xenograft tumor growth in vivo. Moreover, we present compelling evidence demonstrating the ability of sertindole to enhance the cellular response to the chemotherapeutic agent temozolomide both in vitro and in vivo. Additionally, our findings reveal that sertindole effectively suppresses the self-renewal capacity and expression of stemness-associated genes, such as Nanog and Sox2, in glioma tumor cells and glioma stem cells. A mechanistic investigation demonstrated that sertindole enhances the formation of autophagosome–lysosome complexes while concurrently impeding autophagic flux through the inhibition of lysosomal hydrolytic enzymes CTSD and CTSB, ultimately resulting in decreased growth of tumor cells. In conclusion, our findings suggest that sertindole has the potential to develop into a potent antiglioma therapeutic agent.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.41","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50144909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yixun Lu, Guoxiao Liu, Yanjun Wang, Kai Li, Xinxin Xu, Benlong Zhang, Lin Chen, Hongqing Xi, Xinxin Wang
Gastric cancer (GC) is one of the most commonly diagnosed malignancies worldwide. Compelling evidence indicates that circular RNA (circRNA) played critical roles in multiple cancers. However, the role and mechanisms of circRNAs in GC remains unclear. Here we first identified a notably overexpressed circular RNA hsa_circ_0025506 in GC by human circRNA microarray, designated as circGPRC5A(e2). Next, we found circGPRC5A(e2) was overexpressed in GC cell lines and clinical samples as well. Then, we confirmed that circGPRC5A(e2) was primarily located in the cytoplasm of GC cells and colocation phenomenon was observed with miR-665 via fluorescence in situ hybridization. Functionally, Cell Counting Kit-8, 5-Ethynyl-2′-deoxyuridine, clone formation assay, Transwell invasion assay, wound-healing assay, and animal experiments showed that circGPRC5A(e2) promoted GC proliferation, migration, and invasion in vitro, and tumorigenesis and metastasis in vivo. Mechanistically, we showed that circGPRC5A(e2) could serve as miR-665 sponges and facilitate GC growth and metastasis via modulating miR-665/LIM and SH3 protein 1 (LASP1) axis and activating phosphatidylinositol 3-kinase/AKT pathway. Taken together, this study revealed that circGPRC5A(e2) functioned as an oncogene in GC. The circGPRC5A(e2)/miR-665/LASP1 axis revealed by current research might provide novel biomarkers and promising therapeutic targets for GC.
{"title":"Circular RNA circGPRC5A(e2) facilitates gastric cancer progression and metastasis via modulating miR-665/LASP1 and activating PI3K/AKT pathway","authors":"Yixun Lu, Guoxiao Liu, Yanjun Wang, Kai Li, Xinxin Xu, Benlong Zhang, Lin Chen, Hongqing Xi, Xinxin Wang","doi":"10.1002/mog2.39","DOIUrl":"https://doi.org/10.1002/mog2.39","url":null,"abstract":"<p>Gastric cancer (GC) is one of the most commonly diagnosed malignancies worldwide. Compelling evidence indicates that circular RNA (circRNA) played critical roles in multiple cancers. However, the role and mechanisms of circRNAs in GC remains unclear. Here we first identified a notably overexpressed circular RNA hsa_circ_0025506 in GC by human circRNA microarray, designated as circGPRC5A(e2). Next, we found circGPRC5A(e2) was overexpressed in GC cell lines and clinical samples as well. Then, we confirmed that circGPRC5A(e2) was primarily located in the cytoplasm of GC cells and colocation phenomenon was observed with miR-665 via fluorescence in situ hybridization. Functionally, Cell Counting Kit-8, 5-Ethynyl-2′-deoxyuridine, clone formation assay, Transwell invasion assay, wound-healing assay, and animal experiments showed that circGPRC5A(e2) promoted GC proliferation, migration, and invasion in vitro, and tumorigenesis and metastasis in vivo. Mechanistically, we showed that circGPRC5A(e2) could serve as miR-665 sponges and facilitate GC growth and metastasis via modulating miR-665/LIM and SH3 protein 1 (LASP1) axis and activating phosphatidylinositol 3-kinase/AKT pathway. Taken together, this study revealed that circGPRC5A(e2) functioned as an oncogene in GC. The circGPRC5A(e2)/miR-665/LASP1 axis revealed by current research might provide novel biomarkers and promising therapeutic targets for GC.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.39","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50138931","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chimeric antigen receptor (CAR)-T cell therapy is a promising form of cancer immunotherapy that genetically modifies a patient's own T cells to express CARs for the specific recognition and eradication of cancer cells. Unfortunately, unlike the impressive advancements it achieves in hematologic cancer treatment, CAR-T cell therapy has encountered obstacles in treating solid tumors such as high cost, inadequate tumor infiltration, and immunosuppressive tumor microenvironment. Recently, the regional administration of CAR-T cells via hydrogel platforms has been investigated as a potential method to not only promote tumor infiltration, cell expansion, and anticancer efficacy of the CAR-T cells but also provide a multifunctional platform to introduce additional therapeutic agents for achieving potentiated cancer therapy. In this perspective, different design strategies of CAR-T cell delivery hydrogels are introduced. Besides, various types of therapeutic agents incorporated in the hydrogel platforms and diverse hydrogel formulations have been discussed. The current challenges and future research directions on CAR-T cell delivery hydrogels are also proposed. It is hoped that this perspective can help future researchers develop new CAR-T cell delivery hydrogels that can effectively fight against solid tumors.
{"title":"Delivering CAR-T cells into solid tumors via hydrogels","authors":"Shun-Yu Wu, Feng Ji, Bin Xu, Fu-Gen Wu","doi":"10.1002/mog2.40","DOIUrl":"https://doi.org/10.1002/mog2.40","url":null,"abstract":"<p>Chimeric antigen receptor (CAR)-T cell therapy is a promising form of cancer immunotherapy that genetically modifies a patient's own T cells to express CARs for the specific recognition and eradication of cancer cells. Unfortunately, unlike the impressive advancements it achieves in hematologic cancer treatment, CAR-T cell therapy has encountered obstacles in treating solid tumors such as high cost, inadequate tumor infiltration, and immunosuppressive tumor microenvironment. Recently, the regional administration of CAR-T cells via hydrogel platforms has been investigated as a potential method to not only promote tumor infiltration, cell expansion, and anticancer efficacy of the CAR-T cells but also provide a multifunctional platform to introduce additional therapeutic agents for achieving potentiated cancer therapy. In this perspective, different design strategies of CAR-T cell delivery hydrogels are introduced. Besides, various types of therapeutic agents incorporated in the hydrogel platforms and diverse hydrogel formulations have been discussed. The current challenges and future research directions on CAR-T cell delivery hydrogels are also proposed. It is hoped that this perspective can help future researchers develop new CAR-T cell delivery hydrogels that can effectively fight against solid tumors.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.40","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50138932","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In a study recently published in Nature, Liu et al. discovered that lactate directly inhibits SUMO-specific peptidase 1 (SENP1), resulting in the stabilization of anaphase promoting complex (APC) subunit 4 (APC4) SUMOylation, and transient binding of APC/cyclosome (APC/C) and ubiquitin conjugating enzyme E2 C (UBE2C), which promotes the ubiquitination and degradation of cyclin B1 and securin.1 Furthermore, sustained accumulation of lactate was found to counteract the effects of anti-mitotic drugs by inducing mitotic slippage, which ultimately facilitates mitotic exit. This study shed light on a potential mechanism behind the observed high levels of lactate in rapidly dividing cells.
Cancer cells exhibit a unique metabolic phenotype characterized by increased glucose uptake and reliance on aerobic glycolysis to fuel their rapid proliferation. This metabolic shift contributes to lactate accumulation, which is closely associated with cell proliferation; however, the precise mechanism of the latter remains unclear. APC/C is a member of the ubiquitin ligase family that plays a crucial role in regulating the metaphase-to-anaphase transition and mitotic exit by assembling K11-linked ubiquitin chains on substrates such as cyclin B1 and securin.2 A recently published work by Liu et al. uncovered a link between lactate and APC/C activity, and elucidated the significance of this connection in cell cycle and cell proliferation modulation.
To explore the direct effect of elevated lactate levels on the entire proteome, Liu et al. treated native human embryonic kidney cell lysates with 15 or 25 mM l-lactate before conducting thermal proteomic profiling. They observed a significant shift in the thermostability of UBE2C, an E2 enzyme recruited by APC/C upon structural reorganization of its subunits. However, it is unlikely that lactate binds directly to UBE2C because of its low affinity. Moreover, no change in the abundance or posttranslational modification of UBE2C was detected, suggesting that l-lactate might enhance the interaction between UBE2C and APC/C. To verify this hypothesis, cells were first synchronized to pro-metaphase, a period during which APC/C is inhibited due to its interaction with the mitotic checkpoint complex. Subsequently, Liu et al. incubated cell lysates with 15 mM l-lactate and performed immunoprecipitation of APC/C, revealing that l-lactate significantly enhanced the binding between UBE2C and APC/C. In addition, mass spectrometry analysis of APC/C showed a lactate-dependent elevation of SUMO2/3 conjunctions. Previous studies have shown that SUMOylation of APC4 on residues K772 and K798 results in a substantial rearrangement of the WHB domain in APC2, facilitating the binding of UBE2C to APC/C for an efficient APC/C activation.3, 4 To further investigate the role of APC4 SUMOylation in lactate-
在最近发表在《自然》杂志上的一项研究中,刘等人发现乳酸直接抑制SUMO特异性肽酶1(SENP1),导致后期促进复合物(APC)亚基4(APC4)SUMO化的稳定,以及APC/环体(APC/C)和泛素结合酶E2 C(UBE2C)的瞬时结合,其促进细胞周期蛋白B1和securin的泛素化和降解。1此外,发现乳酸的持续积累通过诱导有丝分裂滑移来抵消抗有丝分裂药物的作用,最终促进有丝分裂退出。这项研究揭示了在快速分裂的细胞中观察到的高水平乳酸背后的潜在机制。癌症细胞表现出独特的代谢表型,其特征是葡萄糖摄取增加,并依赖有氧糖酵解来促进其快速增殖。这种代谢变化有助于乳酸的积累,而乳酸的积累与细胞增殖密切相关;然而,后者的确切机制尚不清楚。APC/C是泛素连接酶家族的一员,通过在细胞周期蛋白B1和securin等底物上组装K11连接的泛素链,在调节中后期过渡和有丝分裂退出中发挥着至关重要的作用。2刘等人最近发表的一项工作揭示了乳酸和APC/C活性之间的联系,并阐明了这种联系在细胞周期和细胞增殖调节中的意义。为了探索乳酸水平升高对整个蛋白质组的直接影响,刘等人用15或25 mM l-乳酸,然后进行热蛋白质组学分析。他们观察到UBE2C的热稳定性发生了显著变化,UBE2C是一种在亚基结构重组时被APC/C招募的E2酶。然而,乳酸盐不太可能直接与UBE2C结合,因为其亲和力低。此外,未检测到UBE2C的丰度或翻译后修饰的变化,表明l-乳酸可能增强UBE2C和APC/C之间的相互作用。为了验证这一假设,首先将细胞同步到前中期,在这段时间内,APC/C由于与有丝分裂检查点复合体的相互作用而受到抑制。随后,刘等人用15 mM l-乳酸并进行APC/C的免疫沉淀,揭示l-乳酸显著增强UBE2C和APC/C之间的结合。此外,APC/C的质谱分析显示SUMO2/3结合物的乳酸依赖性升高。先前的研究表明,APC4在残基K772和K798上的SUMO化导致APC2中WHB结构域的实质性重排,促进UBE2C与APC/C的结合以进行有效的APC/C激活。3,4为了进一步研究APC4 SUMO化在乳酸盐介导的UBE2C–APC/C相互作用中的作用,刘等人构建了APC4 K772/798R细胞,其中APC4 SUMO化被消除。他们发现,在这些细胞中,乳酸盐介导的UBE2C和APC/C之间的相互作用完全丧失。刘等人随后研究了l-乳酸影响APC4 SUMO化的机制,并提出乳酸直接靶向SENP1,这是一种对有丝分裂至关重要的去SUMO化酶。支持这一推测的结果表明,删除SENP1概括了乳酸水平升高的影响,包括APC4 SUMO化增加以及UBE2C和APC/C的强结合。此外,增加SENP1缺陷细胞中的乳酸浓度并没有进一步增强APC4 SUMO化。进一步的分析显示,SENP1的活性位点在结构上类似于锌结合口袋,并且乳酸盐取代SENP1的His533侧链以结合锌。此外,SENP1中的Cys535和Asn556对于乳酸对SENP1活性的影响是必需的。乳酸通过与Asn556形成氢键来抑制SENP1活性,从而增强SENP1和锌螯合作用,而在携带SENP1中N556A突变的细胞中,乳酸介导的APC4 SUMO化被阻止(图1A)。为了确定新发现的乳酸功能是否在有丝分裂的调节中发挥广泛作用,刘等人跟踪了同步的HeLa S3和HCT116细胞中的乳酸水平,发现它们在进入有丝分裂时增加。当内源性乳酸达到15 mM,它触发了APC/C的定时重塑和cyclinB1和securin的降解,允许中期到后期的转变和有丝分裂退出。这些发现表明,增殖细胞在进入有丝分裂时自然积累达到APC/C重塑所需浓度的乳酸。抗寄生虫药物可以抑制细胞的快速增殖,并可用于癌症的治疗。诺可唑在中期阻止姐妹染色单体的动粒与有丝分裂纺锤体结合,并通过干扰微管聚合激活纺锤体组装检查点(SAC)。
{"title":"Lactate: A critical regulator of cell proliferation via anaphase promoting complex remodeling","authors":"Qiqing Yang, Long Zhang, Jun Chen","doi":"10.1002/mog2.38","DOIUrl":"https://doi.org/10.1002/mog2.38","url":null,"abstract":"<p>In a study recently published in <i>Nature</i>, Liu et al. discovered that lactate directly inhibits SUMO-specific peptidase 1 (SENP1), resulting in the stabilization of anaphase promoting complex (APC) subunit 4 (APC4) SUMOylation, and transient binding of APC/cyclosome (APC/C) and ubiquitin conjugating enzyme E2 C (UBE2C), which promotes the ubiquitination and degradation of cyclin B1 and securin.<span><sup>1</sup></span> Furthermore, sustained accumulation of lactate was found to counteract the effects of anti-mitotic drugs by inducing mitotic slippage, which ultimately facilitates mitotic exit. This study shed light on a potential mechanism behind the observed high levels of lactate in rapidly dividing cells.</p><p>Cancer cells exhibit a unique metabolic phenotype characterized by increased glucose uptake and reliance on aerobic glycolysis to fuel their rapid proliferation. This metabolic shift contributes to lactate accumulation, which is closely associated with cell proliferation; however, the precise mechanism of the latter remains unclear. APC/C is a member of the ubiquitin ligase family that plays a crucial role in regulating the metaphase-to-anaphase transition and mitotic exit by assembling K11-linked ubiquitin chains on substrates such as cyclin B1 and securin.<span><sup>2</sup></span> A recently published work by Liu et al. uncovered a link between lactate and APC/C activity, and elucidated the significance of this connection in cell cycle and cell proliferation modulation.</p><p>To explore the direct effect of elevated lactate levels on the entire proteome, Liu et al. treated native human embryonic kidney cell lysates with 15 or 25 mM <span>l</span>-lactate before conducting thermal proteomic profiling. They observed a significant shift in the thermostability of UBE2C, an E2 enzyme recruited by APC/C upon structural reorganization of its subunits. However, it is unlikely that lactate binds directly to UBE2C because of its low affinity. Moreover, no change in the abundance or posttranslational modification of UBE2C was detected, suggesting that <span>l</span>-lactate might enhance the interaction between UBE2C and APC/C. To verify this hypothesis, cells were first synchronized to pro-metaphase, a period during which APC/C is inhibited due to its interaction with the mitotic checkpoint complex. Subsequently, Liu et al. incubated cell lysates with 15 mM <span>l</span>-lactate and performed immunoprecipitation of APC/C, revealing that <span>l</span>-lactate significantly enhanced the binding between UBE2C and APC/C. In addition, mass spectrometry analysis of APC/C showed a lactate-dependent elevation of SUMO2/3 conjunctions. Previous studies have shown that SUMOylation of APC4 on residues K772 and K798 results in a substantial rearrangement of the WHB domain in APC2, facilitating the binding of UBE2C to APC/C for an efficient APC/C activation.<span><sup>3, 4</sup></span> To further investigate the role of APC4 SUMOylation in lactate-","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.38","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50121884","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pyroptosis is an immunogenic cell death and would trigger robust antitumor immunity. However, due to cytoxicity of pyroptosis executors, Gasdermin family proteins, it is indispensable to construct tumor-specific vectors. Here, we report the development of a novel vector named lipid-coated poly(lactide-co-glycolide) (PLGA) nanoparticles coloaded with Gasdermin E expressing plasmid DNA (GSDME-pDNA) with a heat-inducible mouse heat shock protein 70 (mHSP70) as the promoter and a photosensitizer indocyanine green (ICG) to activate the mHSP70 element. The cellular internalization and transfection rate of the vector were remarkably enhanced by photothermal treatment. And the mHSP70 promoter further improved the gene transfection rate for about 15-fold. With the combination of oxaliplatin (OXA), the mechanism switch between apoptosis and pyroptosis was fulfilled by cleavage of GSDME through activated caspase-3, which promoted damage-associated molecular patterns (DAMPs) release and increased the infiltration of immune cells at the tumor site. This combination strategy not only prominently inhibited the growth of the treated tumor, but also exhibited a lethal effect on the distal tumors. Besides, mouse colon cancer cell CT26 overexpressing GSDME after OXA treatment had the potential to be the preventive tumor vaccine. This study provides a novel thought and feasible method for the clinical treatment of colon cancer.
{"title":"Gasdermin E plasmid DNA/indocyanine green coloaded hybrid nanoparticles with spatiotemporal controllability to induce pyroptosis for colon cancer treatment","authors":"Ailing Jiang, Mao Wang, Huimin Liu, Simeng Liu, Xiaoshuang Song, Yu Zou, Yuchuan Deng, Qin Qin, Yiran Song, Yu Zheng","doi":"10.1002/mog2.33","DOIUrl":"https://doi.org/10.1002/mog2.33","url":null,"abstract":"<p>Pyroptosis is an immunogenic cell death and would trigger robust antitumor immunity. However, due to cytoxicity of pyroptosis executors, Gasdermin family proteins, it is indispensable to construct tumor-specific vectors. Here, we report the development of a novel vector named lipid-coated poly(lactide-co-glycolide) (PLGA) nanoparticles coloaded with Gasdermin E expressing plasmid DNA (GSDME-pDNA) with a heat-inducible mouse heat shock protein 70 (mHSP70) as the promoter and a photosensitizer indocyanine green (ICG) to activate the mHSP70 element. The cellular internalization and transfection rate of the vector were remarkably enhanced by photothermal treatment. And the mHSP70 promoter further improved the gene transfection rate for about 15-fold. With the combination of oxaliplatin (OXA), the mechanism switch between apoptosis and pyroptosis was fulfilled by cleavage of GSDME through activated caspase-3, which promoted damage-associated molecular patterns (DAMPs) release and increased the infiltration of immune cells at the tumor site. This combination strategy not only prominently inhibited the growth of the treated tumor, but also exhibited a lethal effect on the distal tumors. Besides, mouse colon cancer cell CT26 overexpressing GSDME after OXA treatment had the potential to be the preventive tumor vaccine. This study provides a novel thought and feasible method for the clinical treatment of colon cancer.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.33","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50117711","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lijing Du, Shasha Li, Xue Xiao, Jin Li, Yuanfang Sun, Shuai Ji, Huizi Jin, Zhaolai Hua, Juming Ma, Xi Wang, Shikai Yan
Gastric cancer (GC) faces a great challenge in clinical diagnosis, that it often is detected at advanced stages and there is a loss of optimum time for treatment. Thus, it is necessary to develop effective strategies for diagnosis of GC. In this study, 82 participants were enrolled, including 50 chronic superficial gastritis (CSG) patients, 7 early gastric cancer (EGC), and 25 advanced gastric cancer (AGC) ones. Metabolites profiling on patient plasma was performed. Furthermore, the proposed biomarkers were used to create random forest models, in which discrimination efficiency and accuracy were ascertained by receiver operating characteristic (ROC) curve analysis. l-carnitine, l-proline, pyruvaldehyde, phosphatidylcholines (PC) (14:0/18:0), lysophosphatidylcholine (14:0) (LysoPC 14:0), lysinoalanine were defined as the potential biomarker panel for the diagnosis among CSG and EGC patients. Compared with EGC patients, PC(O-18:0/0:0) and LysoPC(20:4(5Z,8Z,11Z,14Z)) were found to be upregulated in AGC patients, whereasl-proline, l-valine, adrenic acid, and pyruvaldehyde downregulated. Pathway analysis revealed several metabolism disorders, involving amino acids and lipid metabolism. ROC analysis demonstrated a high diagnostic performance in disease diagnosis between CSG and GC. The above results indicate that the biomarker panels are sensitive to early diagnosis of GC disease, which is expected to be a promising diagnostic tool for disease stratification studies.
{"title":"Metabolomic profiling of plasma reveals potential biomarkers for screening and early diagnosis of gastric cancer and precancerous stages","authors":"Lijing Du, Shasha Li, Xue Xiao, Jin Li, Yuanfang Sun, Shuai Ji, Huizi Jin, Zhaolai Hua, Juming Ma, Xi Wang, Shikai Yan","doi":"10.1002/mog2.32","DOIUrl":"https://doi.org/10.1002/mog2.32","url":null,"abstract":"<p>Gastric cancer (GC) faces a great challenge in clinical diagnosis, that it often is detected at advanced stages and there is a loss of optimum time for treatment. Thus, it is necessary to develop effective strategies for diagnosis of GC. In this study, 82 participants were enrolled, including 50 chronic superficial gastritis (CSG) patients, 7 early gastric cancer (EGC), and 25 advanced gastric cancer (AGC) ones. Metabolites profiling on patient plasma was performed. Furthermore, the proposed biomarkers were used to create random forest models, in which discrimination efficiency and accuracy were ascertained by receiver operating characteristic (ROC) curve analysis. <span>l</span>-carnitine, <span>l</span>-proline, pyruvaldehyde, phosphatidylcholines (PC) (14:0/18:0), lysophosphatidylcholine (14:0) (LysoPC 14:0), lysinoalanine were defined as the potential biomarker panel for the diagnosis among CSG and EGC patients. Compared with EGC patients, PC(O-18:0/0:0) and LysoPC(20:4(5Z,8Z,11Z,14Z)) were found to be upregulated in AGC patients, whereas<span>l</span>-proline, <span>l</span>-valine, adrenic acid, and pyruvaldehyde downregulated. Pathway analysis revealed several metabolism disorders, involving amino acids and lipid metabolism. ROC analysis demonstrated a high diagnostic performance in disease diagnosis between CSG and GC. The above results indicate that the biomarker panels are sensitive to early diagnosis of GC disease, which is expected to be a promising diagnostic tool for disease stratification studies.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-05-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.32","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50142201","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}