Pub Date : 2024-09-14DOI: 10.1016/j.tranon.2024.102121
Nihat Dilsiz
Exosomes are small, round vesicles in the 30 and 120 nm diameter range released by all living cell types. Exosomes play many essential functions in intercellular communication and tissue crosstalk in the human body. They can potentially be used as strong biomarkers and therapeutic agents for early diagnosis, therapy response, and prognosis of different diseases. The main requirements for exosomal large-scale clinical practice application are rapid, easy, high-yield, high purity, characterization, safety, low cost, and therapeutic efficacy. Depending on the sample types, environmental insults, and exosome quantity, exosomes can be isolated from various sources, including body fluids, solid tissues, and cell culture medium using different procedures. This study comprehensively analyzed the current research progress in exosome isolation and characterization strategies along with their advantages and disadvantages. The provided information will make it easier to select exosome separation methods based on the types of biological samples available, and it will facilitate the use of exosomes in translational and clinical research, particularly in cancer.
Lay abstract Exosomes have recently received much attention due to their potential to function as biomarkers and novel therapeutic agents for early diagnosis, therapeutic response, and prognosis in various diseases. This review summarizes many approaches for isolating and characterizing exosomes, focusing on developing technologies, and provides an in-depth comparison and analysis of each method, including its principles, advantages, and limitations.
{"title":"A comprehensive review on recent advances in exosome isolation and characterization: Toward clinical applications","authors":"Nihat Dilsiz","doi":"10.1016/j.tranon.2024.102121","DOIUrl":"10.1016/j.tranon.2024.102121","url":null,"abstract":"<div><p>Exosomes are small, round vesicles in the 30 and 120 nm diameter range released by all living cell types. Exosomes play many essential functions in intercellular communication and tissue crosstalk in the human body. They can potentially be used as strong biomarkers and therapeutic agents for early diagnosis, therapy response, and prognosis of different diseases. The main requirements for exosomal large-scale clinical practice application are rapid, easy, high-yield, high purity, characterization, safety, low cost, and therapeutic efficacy. Depending on the sample types, environmental insults, and exosome quantity, exosomes can be isolated from various sources, including body fluids, solid tissues, and cell culture medium using different procedures. This study comprehensively analyzed the current research progress in exosome isolation and characterization strategies along with their advantages and disadvantages. The provided information will make it easier to select exosome separation methods based on the types of biological samples available, and it will facilitate the use of exosomes in translational and clinical research, particularly in cancer.</p><p><strong>Lay abstract</strong> Exosomes have recently received much attention due to their potential to function as biomarkers and novel therapeutic agents for early diagnosis, therapeutic response, and prognosis in various diseases. This review summarizes many approaches for isolating and characterizing exosomes, focusing on developing technologies, and provides an in-depth comparison and analysis of each method, including its principles, advantages, and limitations.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"50 ","pages":"Article 102121"},"PeriodicalIF":5.0,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002481/pdfft?md5=aa35800e03a6f6c513fc2cd66d9eeb2e&pid=1-s2.0-S1936523324002481-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142229077","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-12DOI: 10.1016/j.tranon.2024.102119
Erika Nakatsuka , Lijun Tan , Brianna Cunneen , Caroline Foster , Yu Leo Lei , Karen McLean
While poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) have improved the prognosis of ovarian high-grade serous carcinoma (HGSC) tumors that are homologous recombination (HR) deficient (HRD), new therapeutic strategies are needed for tumors that are HR proficient (HRP) because they demonstrate greater resistance to current treatments and thus have poorer clinical outcomes. Additionally, clinical precautionary statements regarding potential risks associated with PARPi, such as myelodysplastic syndrome, highlight the need for combinatorial approaches that can lessen the dose and duration of PARPi treatment to reduce toxicities. Here, we evaluated DNA double-strand damage repair pathways in HRD and HRP ovarian cancer cell lines and found that in HRD cell lines, PARPi therapy reduced non-homologous end joining (NHEJ)-mediated repair, specifically due to decreased theta-mediated end-joining. The combination of PARPi with ATM serine/threonine kinase inhibitor (ATMi) suppressed both NHEJ and HR pathways in HRD and HRP cell lines, with synergistic increases in apoptosis and decreases in cell viability and colony formation. Interestingly, PARPi plus ATMi also decreased NF-κB p65 phosphorylation, which was not observed when PARPi was combined with inhibition of the ATR kinase (ATRi). These findings indicate that PARPi plus ATMi is a promising strategy for HGSC independent of underlying tumor HR status.
{"title":"Characterization of DNA damage repair pathway utilization in high-grade serous ovarian cancers yields rational therapeutic approaches","authors":"Erika Nakatsuka , Lijun Tan , Brianna Cunneen , Caroline Foster , Yu Leo Lei , Karen McLean","doi":"10.1016/j.tranon.2024.102119","DOIUrl":"10.1016/j.tranon.2024.102119","url":null,"abstract":"<div><p>While poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) have improved the prognosis of ovarian high-grade serous carcinoma (HGSC) tumors that are homologous recombination (HR) deficient (HRD), new therapeutic strategies are needed for tumors that are HR proficient (HRP) because they demonstrate greater resistance to current treatments and thus have poorer clinical outcomes. Additionally, clinical precautionary statements regarding potential risks associated with PARPi, such as myelodysplastic syndrome, highlight the need for combinatorial approaches that can lessen the dose and duration of PARPi treatment to reduce toxicities. Here, we evaluated DNA double-strand damage repair pathways in HRD and HRP ovarian cancer cell lines and found that in HRD cell lines, PARPi therapy reduced non-homologous end joining (NHEJ)-mediated repair, specifically due to decreased theta-mediated end-joining. The combination of PARPi with ATM serine/threonine kinase inhibitor (ATMi) suppressed both NHEJ and HR pathways in HRD and HRP cell lines, with synergistic increases in apoptosis and decreases in cell viability and colony formation. Interestingly, PARPi plus ATMi also decreased NF-κB p65 phosphorylation, which was not observed when PARPi was combined with inhibition of the ATR kinase (ATRi). These findings indicate that PARPi plus ATMi is a promising strategy for HGSC independent of underlying tumor HR status.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"50 ","pages":"Article 102119"},"PeriodicalIF":5.0,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002468/pdfft?md5=580e5f10a5b02eb990d448b737acd62a&pid=1-s2.0-S1936523324002468-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142167867","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-11DOI: 10.1016/j.tranon.2024.102118
Xiao-Liang Wang , Xiangfei He , Tong Gao , Xinyu Zhou , Zobeida Cruz-Monserrate , Allan Tsung , Jianjie Ma , Chuanxi Cai
Pancreatic ductal adenocarcinoma (PDAC) poses a formidable challenge in oncology due to its limited treatment options and poor long-term survival rates. Our previous work identified MG53, a member of the tripartite motif family protein (TRIM72), as a key player in tissue repair with potential applications in regenerative medicine. Despite the focus on MG53’s cytosolic functions, its nuclear role in suppressing pancreatic cancer remains unknown. Through orthotopic and subcutaneous transplantation studies in mice, we observed enhanced tumor growth in MG53-deficient mice compared to wild-type counterparts. The overexpression of KIF11, a motor protein crucial for cell mitosis regulation, has been linked to the aggressive proliferation of pancreatic cancer cells. Confocal imaging confirmed MG53′s presence in the nucleus of human pancreatic cancer cells, while functional assays demonstrated its impact on KIF11 expression and subsequent cell proliferation. Mechanistically, we revealed MG53′s transcriptional control over KIF11, leading to cell cycle arrest. Our findings position MG53 as a promising tumor suppressor in PDAC, offering a novel avenue for therapeutic intervention by regulating KIF11 expression.
{"title":"MG53 suppresses tumor growth via transcriptional inhibition of KIF11 in pancreatic cancer","authors":"Xiao-Liang Wang , Xiangfei He , Tong Gao , Xinyu Zhou , Zobeida Cruz-Monserrate , Allan Tsung , Jianjie Ma , Chuanxi Cai","doi":"10.1016/j.tranon.2024.102118","DOIUrl":"10.1016/j.tranon.2024.102118","url":null,"abstract":"<div><p>Pancreatic ductal adenocarcinoma (PDAC) poses a formidable challenge in oncology due to its limited treatment options and poor long-term survival rates. Our previous work identified MG53, a member of the tripartite motif family protein (TRIM72), as a key player in tissue repair with potential applications in regenerative medicine. Despite the focus on MG53’s cytosolic functions, its nuclear role in suppressing pancreatic cancer remains unknown. Through orthotopic and subcutaneous transplantation studies in mice, we observed enhanced tumor growth in MG53-deficient mice compared to wild-type counterparts. The overexpression of KIF11, a motor protein crucial for cell mitosis regulation, has been linked to the aggressive proliferation of pancreatic cancer cells. Confocal imaging confirmed MG53′s presence in the nucleus of human pancreatic cancer cells, while functional assays demonstrated its impact on KIF11 expression and subsequent cell proliferation. Mechanistically, we revealed MG53′s transcriptional control over KIF11, leading to cell cycle arrest. Our findings position MG53 as a promising tumor suppressor in PDAC, offering a novel avenue for therapeutic intervention by regulating KIF11 expression.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"50 ","pages":"Article 102118"},"PeriodicalIF":5.0,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002456/pdfft?md5=df16f9329ec363216f0fd7364e72d4ae&pid=1-s2.0-S1936523324002456-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142167152","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-09DOI: 10.1016/j.tranon.2024.102111
Miaomiao Zhang , Haiting Wang , Meng Wang , Haoliang Zhang , Huizhong Li , Ping Ma , Junnian Zheng , Gang Wang , Shibao Li
Erythropoietin-producing hepatocyte receptor A2 (EphA2) is an attractive target for immunotherapy due to its high expression in a variety of solid tumors including prostate cancer. Among various types of immunotherapeutics, chimeric antigen receptor T (CAR-T) cell therapy has made promising progress in hematological and solid tumors. Here, we detected the expression of EphA2 in prostate cancer cells and developed a second-generation CAR targeting EphA2 with CD28 as a co-stimulatory receptor to explore its tumor suppressive potential for prostate cancer in vitro and in vivo. EphA2 was highly expressed on the surface of PC3 and DU145 cells. EphA2 CART cells effectively inhibited prostate cancer growth in an antigen-dependent manner in vitro and in vivo. In addition, tumor cells could stimulate the proliferation of CAR-T cells and the release of cytokine IFN-γ in vitro. These findings shed light on EphA2 as a potential target for prostate cancer, promising EphA2 specific CAR-T cells for the treatment of prostate cancer.
{"title":"EphA2 specific chimeric antigen receptor engineered T cells for the treatment of prostate cancer","authors":"Miaomiao Zhang , Haiting Wang , Meng Wang , Haoliang Zhang , Huizhong Li , Ping Ma , Junnian Zheng , Gang Wang , Shibao Li","doi":"10.1016/j.tranon.2024.102111","DOIUrl":"10.1016/j.tranon.2024.102111","url":null,"abstract":"<div><p>Erythropoietin-producing hepatocyte receptor A2 (EphA2) is an attractive target for immunotherapy due to its high expression in a variety of solid tumors including prostate cancer. Among various types of immunotherapeutics, chimeric antigen receptor T (CAR-T) cell therapy has made promising progress in hematological and solid tumors. Here, we detected the expression of EphA2 in prostate cancer cells and developed a second-generation CAR targeting EphA2 with CD28 as a co-stimulatory receptor to explore its tumor suppressive potential for prostate cancer in vitro <em>and</em> in vivo. EphA2 was highly expressed on the surface of PC3 and DU145 cells. EphA2 CART cells effectively inhibited prostate cancer growth in an antigen-dependent manner in vitro <em>and</em> in vivo. In addition, tumor cells could stimulate the proliferation of CAR-T cells and the release of cytokine IFN-γ in vitro. These findings shed light on EphA2 as a potential target for prostate cancer, promising EphA2 specific CAR-T cells for the treatment of prostate cancer.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"50 ","pages":"Article 102111"},"PeriodicalIF":5.0,"publicationDate":"2024-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002389/pdfft?md5=80d564b9c6b4228dc69096825ed5d559&pid=1-s2.0-S1936523324002389-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142157885","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-05DOI: 10.1016/j.tranon.2024.102117
Shuangshuang Mao , Yuanyuan Zhao , Huihua Xiong , Chen Gong
Regulated cell death (RCD) has been documented to have great potentials for discovering novel biomarkers and therapeutic targets in malignancies. But its role and clinical value in HR+/HER2- breast cancer, the most common subtype of breast cancer, are obscure. In this study, we comprehensively explored 12 types of RCD patterns and found extensive mutations and dysregulations of RCD genes in HR+/HER2- breast cancer. A prognostic RCD scoring system (CDScore) based on six critical genes (LEF1, SLC7A11, SFRP1, IGFBP6, CXCL2, STXBP1) was constructed, in which a high CDScore predicts poor prognosis. The expressions and prognostic value of LEF1 and SFRP1were also validated in our tissue microarrays. The nomogram established basing on CDScore, age and TNM stage performed satisfactory in predicting overall survival, with an area under the ROC curve of 0.89, 0.82 and 0.8 in predicting 1-year, 3-year and 5-year overall survival rates, respectively. Furthermore, CDScore was identified to be correlated with tumor microenvironments and immune checkpoints by excavation of bulk and single-cell sequencing data. Patients in CDScore high group might be resistant to standard chemotherapy and target therapy. Our results underlined the potential effects and importance of RCD in HR+/HER2- breast cancer and provided novel biomarkers and therapeutic targets for HR+/HER2- breast cancer patients.
{"title":"Excavating regulated cell death signatures to predict prognosis, tumor microenvironment and therapeutic response in HR+/HER2- breast cancer","authors":"Shuangshuang Mao , Yuanyuan Zhao , Huihua Xiong , Chen Gong","doi":"10.1016/j.tranon.2024.102117","DOIUrl":"10.1016/j.tranon.2024.102117","url":null,"abstract":"<div><p>Regulated cell death (RCD) has been documented to have great potentials for discovering novel biomarkers and therapeutic targets in malignancies. But its role and clinical value in HR+/HER2- breast cancer, the most common subtype of breast cancer, are obscure. In this study, we comprehensively explored 12 types of RCD patterns and found extensive mutations and dysregulations of RCD genes in HR+/HER2- breast cancer. A prognostic RCD scoring system (CDScore) based on six critical genes (LEF1, SLC7A11, SFRP1, IGFBP6, CXCL2, STXBP1) was constructed, in which a high CDScore predicts poor prognosis. The expressions and prognostic value of LEF1 and SFRP1were also validated in our tissue microarrays. The nomogram established basing on CDScore, age and TNM stage performed satisfactory in predicting overall survival, with an area under the ROC curve of 0.89, 0.82 and 0.8 in predicting 1-year, 3-year and 5-year overall survival rates, respectively. Furthermore, CDScore was identified to be correlated with tumor microenvironments and immune checkpoints by excavation of bulk and single-cell sequencing data. Patients in CDScore high group might be resistant to standard chemotherapy and target therapy. Our results underlined the potential effects and importance of RCD in HR+/HER2- breast cancer and provided novel biomarkers and therapeutic targets for HR+/HER2- breast cancer patients.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"50 ","pages":"Article 102117"},"PeriodicalIF":5.0,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002444/pdfft?md5=021f6e226d0758f7b6b439faf1766bfc&pid=1-s2.0-S1936523324002444-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142146302","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-03DOI: 10.1016/j.tranon.2024.102116
Mareike Mannewitz , Thomas Kolben , Carolin Perleberg , Sarah Meister , Laura Hahn , Sophie Mitter , Elisa Schmoeckel , Sven Mahner , Stefanie Corradini , Fabian Trillsch , Mirjana Kessler , Udo Jeschke , Susanne Beyer
Objectives
The chemokine CCL22 is recognized for recruiting immunosuppressive regulatory T-cells (Treg) that contribute to disease progression in various tumor entities helping them to evade the host immune response. Our study aims to identify the expressing cell types and to evaluate the prognostic significance of CCL22 secretion and its association with Treg invasion in endometrial cancer (EC), an immunogenic cancer.
Methods
Specimens from 275 patients with EC and 28 healthy controls were screened immunohistochemically for CCL22. Immunofluorescence double-staining for CCL22 and different immune cell markers was performed. In vitro regulation of CCL22-expression was examined in EC cell lines (Ishikawa+, RL95–2) and human PBMCs in coculture settings via qPCR and ELISA.
Results
Elevated CCL22 staining in tumor cells and CCL22-positive M1-macrophages in tumordistant areas were significantly associated with increased overall survival (OS). Conversely, high, secretory-appearing staining in the peritumoral and intratumoral stroma correlated with reduced OS. Although the analysis of the in vitro coculture model of epithelial tumor- and immune cells revealed PBMCs as the primary source of CCL22, we could confirm expression of the chemokine also in the EC epithelial cells.
Conclusion
Our study suggests that CCL22 in EC is associated with OS, dependent on its location and the cell type producing it. Intracellular upregulation and extracellular secretion must be considered separately when investigating CCL22 expressing cell types in EC. These results may provide evidence for CCL22-mediated Treg recruitment in EC as a potential future therapeutic target.
{"title":"CCL22 as an independent prognostic factor in endometrial cancer patients","authors":"Mareike Mannewitz , Thomas Kolben , Carolin Perleberg , Sarah Meister , Laura Hahn , Sophie Mitter , Elisa Schmoeckel , Sven Mahner , Stefanie Corradini , Fabian Trillsch , Mirjana Kessler , Udo Jeschke , Susanne Beyer","doi":"10.1016/j.tranon.2024.102116","DOIUrl":"10.1016/j.tranon.2024.102116","url":null,"abstract":"<div><h3>Objectives</h3><p>The chemokine CCL22 is recognized for recruiting immunosuppressive regulatory T-cells (Treg) that contribute to disease progression in various tumor entities helping them to evade the host immune response. Our study aims to identify the expressing cell types and to evaluate the prognostic significance of CCL22 secretion and its association with Treg invasion in endometrial cancer (EC), an immunogenic cancer.</p></div><div><h3>Methods</h3><p>Specimens from 275 patients with EC and 28 healthy controls were screened immunohistochemically for CCL22. Immunofluorescence double-staining for CCL22 and different immune cell markers was performed. In vitro regulation of CCL22-expression was examined in EC cell lines (Ishikawa+, RL95–2) and human PBMCs in coculture settings via qPCR and ELISA.</p></div><div><h3>Results</h3><p>Elevated CCL22 staining in tumor cells and CCL22-positive M1-macrophages in tumordistant areas were significantly associated with increased overall survival (OS). Conversely, high, secretory-appearing staining in the peritumoral and intratumoral stroma correlated with reduced OS. Although the analysis of the in vitro coculture model of epithelial tumor- and immune cells revealed PBMCs as the primary source of CCL22, we could confirm expression of the chemokine also in the EC epithelial cells.</p></div><div><h3>Conclusion</h3><p>Our study suggests that CCL22 in EC is associated with OS, dependent on its location and the cell type producing it. Intracellular upregulation and extracellular secretion must be considered separately when investigating CCL22 expressing cell types in EC. These results may provide evidence for CCL22-mediated Treg recruitment in EC as a potential future therapeutic target.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"50 ","pages":"Article 102116"},"PeriodicalIF":5.0,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002432/pdfft?md5=b29186b49bb5434ee8561a1198172e61&pid=1-s2.0-S1936523324002432-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142128274","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-02DOI: 10.1016/j.tranon.2024.102112
Changbin Chu , Shangjing Liu , Zhiting He , Mingjun Wu , Jing xia , Hongxiang Zeng , Wenhua Xie , Rui Cheng , Xueya Zhao , Xi Li
Background
Clear cell renal cell carcinoma (ccRCC) is a serious threat to human life. It is very important to clarify the pathogenesis of ccRCC. In this study we evaluated the clinical value of HADH and explored its role and mechanism in the malignant progression of ccRCC.
Methods
HADH expression and its relationship with prognosis were analyzed using bioinformatics database. RT-PCR, Western blot and immunohistochemistry were used to examine the expression of HADH in ccRCC tissues and tissue microarrays. To examine the cell proliferation, apoptosis, migration and invasion ability, ccRCC cells with HADH overexpressed were constructed. Xenograft experiments were performed to determine the role of HADH. Non-target metabolomics was applied to explore the potential metabolic pathway by which HADH inhibited ccRCC progression. Plasmid pcDNA3.1-NRF2 was used to confirm whether HADH inhibited the process of ccRCC cells through NRF2-related glutathione (GSH) synthesis.
Results
Bioinformatics database analysis showed that HADH expression was significantly decreased in ccRCC tissues, and its low expression predicted a poor prognosis. Both ccRCC tissues and tissue microarrays exhibited a significantly decreased HADH level compared with adjacent normal renal tissues. HADH overexpression inhibited the malignant behaviors of ccRCC cells. Furthermore, HADH overexpression attenuated GSH synthesis and induced oxidative stress damage. Exogenously increased NRF2 effectively attenuated the inhibitive effect of HADH overexpression on ccRCC cells.
Conclusion
Our data revealed that HADH suppressed the malignant behaviors of ccRCC cells by attenuating GSH synthesis through inhibition of NRF2 nuclear translocation, and HADH might be a novel therapeutic target for ccRCC treatment.
{"title":"HADH suppresses clear cell renal cell carcinoma progression through reduced NRF2-dependent glutathione synthesis","authors":"Changbin Chu , Shangjing Liu , Zhiting He , Mingjun Wu , Jing xia , Hongxiang Zeng , Wenhua Xie , Rui Cheng , Xueya Zhao , Xi Li","doi":"10.1016/j.tranon.2024.102112","DOIUrl":"10.1016/j.tranon.2024.102112","url":null,"abstract":"<div><h3>Background</h3><p>Clear cell renal cell carcinoma (ccRCC) is a serious threat to human life. It is very important to clarify the pathogenesis of ccRCC. In this study we evaluated the clinical value of HADH and explored its role and mechanism in the malignant progression of ccRCC.</p></div><div><h3>Methods</h3><p>HADH expression and its relationship with prognosis were analyzed using bioinformatics database. RT-PCR, Western blot and immunohistochemistry were used to examine the expression of HADH in ccRCC tissues and tissue microarrays. To examine the cell proliferation, apoptosis, migration and invasion ability, ccRCC cells with HADH overexpressed were constructed. Xenograft experiments were performed to determine the role of HADH. Non-target metabolomics was applied to explore the potential metabolic pathway by which HADH inhibited ccRCC progression. Plasmid pcDNA3.1-NRF2 was used to confirm whether HADH inhibited the process of ccRCC cells through NRF2-related glutathione (GSH) synthesis.</p></div><div><h3>Results</h3><p>Bioinformatics database analysis showed that HADH expression was significantly decreased in ccRCC tissues, and its low expression predicted a poor prognosis. Both ccRCC tissues and tissue microarrays exhibited a significantly decreased HADH level compared with adjacent normal renal tissues. HADH overexpression inhibited the malignant behaviors of ccRCC cells. Furthermore, HADH overexpression attenuated GSH synthesis and induced oxidative stress damage. Exogenously increased NRF2 effectively attenuated the inhibitive effect of HADH overexpression on ccRCC cells.</p></div><div><h3>Conclusion</h3><p>Our data revealed that HADH suppressed the malignant behaviors of ccRCC cells by attenuating GSH synthesis through inhibition of NRF2 nuclear translocation, and HADH might be a novel therapeutic target for ccRCC treatment.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102112"},"PeriodicalIF":5.0,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002390/pdfft?md5=7e4a4d92f8790a91cdddff8088f99cb9&pid=1-s2.0-S1936523324002390-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142122963","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-01DOI: 10.1016/j.tranon.2024.102076
Xuedong Wang , Qiwei Jian , Ziyun Zhang , Juan Gu , Xinping Wang , Yueping Wang
Objectives
Triple-negative breast cancer (TNBC) is the deadliest subtype of breast cancer (BC). Tumor-derived extracellular vesicles (EVs) trigger tumor progression by promoting M2 polarization. Some lncRNAs can be encapsulated into EVs for intercellular communication. Herein, we investigated the mechanism of TNBC-derived EV-shuttled lncRNA MALAT1 on macrophage polarization/tumorigenesis.
Methods
BC-associated targeted EV-derived lncRNAs were screened. Tumor tissues/tissues adjacent to cancer of TNBC patients, and blood samples of all subjects were collected. MALAT1/POSTN mRNA levels in tumor tissues/tissues adjacent to cancer, and MALAT1 expression in EVs and its correlation with TNBC patient overall survival were assessed by RT-qPCR/Kaplan-Meier survival analysis/log-rank test. TNBC patient M2 infiltration was detected by flow cytometry. MALAT1/POSTN levels in EVs/macrophages were regulated by transfection. Hippo/YAP activation was determined by Western blot. Nude mouse xenograft model was established and metastasis was detected by H&E staining.
Results
MALAT1/POSTN were up-regulated and correlated with M2 infiltration/poor prognosis in TNBC patients. TNBC-derived EVs induced M2 polarization. MALAT1 was highly expressed in TNBC-derived EVs and could be transferred to macrophages via EVs to induce M2 polarization. POSTN overexpression diminished the inhibitory effect of MALAT1 knockdown on M2 markers. EVs activated the Hippo/YAP pathway in macrophages. The Hippo/YAP pathway inhibition abrogated the effect of POSTN overexpression on M2 marker expression. TNBC-EV-derived MALAT1 facilitated M2 polarization, and thus promoting occurrence and metastasis of TNBC in vitro and in vivo.
Conclusions
TNBC-EV-derived MALAT1 activated the Hippo/YAP axis by up-regulating POSTN, thereby inducing M2 polarization to promote TNBC occurrence and metastasis in vivo.
目的三阴性乳腺癌(TNBC)是乳腺癌(BC)中最致命的亚型。肿瘤衍生的细胞外囊泡(EVs)通过促进M2极化引发肿瘤进展。一些lncRNA可被包裹到EVs中进行细胞间通信。在此,我们研究了TNBC衍生的EV-shuttled lncRNA MALAT1对巨噬细胞极化/肿瘤发生的机制。收集 TNBC 患者的肿瘤组织/癌旁组织以及所有受试者的血液样本。通过RT-qPCR/Kaplan-Meier生存分析/log-rank检验评估了肿瘤组织/癌症邻近组织中的MALAT1/POSTN mRNA水平、EV中的MALAT1表达及其与TNBC患者总生存期的相关性。通过流式细胞术检测TNBC患者的M2浸润。通过转染调节EVs/巨噬细胞中的MALAT1/POSTN水平。通过 Western 印迹检测 Hippo/YAP 激活情况。结果MALAT1/POSTN上调并与TNBC患者的M2浸润/预后不良相关。TNBC衍生的EV诱导M2极化。MALAT1在TNBC衍生的EVs中高表达,并可通过EVs转移到巨噬细胞中诱导M2极化。POSTN的过表达削弱了MALAT1敲除对M2标记物的抑制作用。EVs激活了巨噬细胞中的Hippo/YAP通路。抑制Hippo/YAP通路可减轻POSTN过表达对M2标记表达的影响。结论TNBC-EV衍生的MALAT1通过上调POSTN激活Hippo/YAP轴,从而诱导M2极化,促进TNBC在体内的发生和转移。
{"title":"Effect of tumor-derived extracellular vesicle-shuttled lncRNA MALAT1 on proliferation, invasion and metastasis of triple-negative breast cancer by regulating macrophage M2 polarization via the POSTN/Hippo/YAP axis","authors":"Xuedong Wang , Qiwei Jian , Ziyun Zhang , Juan Gu , Xinping Wang , Yueping Wang","doi":"10.1016/j.tranon.2024.102076","DOIUrl":"10.1016/j.tranon.2024.102076","url":null,"abstract":"<div><h3>Objectives</h3><p>Triple-negative breast cancer (TNBC) is the deadliest subtype of breast cancer (BC). Tumor-derived extracellular vesicles (EVs) trigger tumor progression by promoting M2 polarization. Some lncRNAs can be encapsulated into EVs for intercellular communication. Herein, we investigated the mechanism of TNBC-derived EV-shuttled lncRNA MALAT1 on macrophage polarization/tumorigenesis.</p></div><div><h3>Methods</h3><p>BC-associated targeted EV-derived lncRNAs were screened. Tumor tissues/tissues adjacent to cancer of TNBC patients, and blood samples of all subjects were collected. MALAT1/POSTN mRNA levels in tumor tissues/tissues adjacent to cancer, and MALAT1 expression in EVs and its correlation with TNBC patient overall survival were assessed by RT-qPCR/Kaplan-Meier survival analysis/log-rank test. TNBC patient M2 infiltration was detected by flow cytometry. MALAT1/POSTN levels in EVs/macrophages were regulated by transfection. Hippo/YAP activation was determined by Western blot. Nude mouse xenograft model was established and metastasis was detected by H&E staining.</p></div><div><h3>Results</h3><p>MALAT1/POSTN were up-regulated and correlated with M2 infiltration/poor prognosis in TNBC patients. TNBC-derived EVs induced M2 polarization. MALAT1 was highly expressed in TNBC-derived EVs and could be transferred to macrophages via EVs to induce M2 polarization. POSTN overexpression diminished the inhibitory effect of MALAT1 knockdown on M2 markers. EVs activated the Hippo/YAP pathway in macrophages. The Hippo/YAP pathway inhibition abrogated the effect of POSTN overexpression on M2 marker expression. TNBC-EV-derived MALAT1 facilitated M2 polarization, and thus promoting occurrence and metastasis of TNBC <em>in vitro</em> and <em>in vivo</em>.</p></div><div><h3>Conclusions</h3><p>TNBC-EV-derived MALAT1 activated the Hippo/YAP axis by up-regulating POSTN, thereby inducing M2 polarization to promote TNBC occurrence and metastasis <em>in vivo</em>.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102076"},"PeriodicalIF":5.0,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002031/pdfft?md5=6f57ccde0c22a156c7d194a23faa9804&pid=1-s2.0-S1936523324002031-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142117427","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-31DOI: 10.1016/j.tranon.2024.102109
Jingyu An , Roma Kurilov , Teresa Peccerella , Frank Bergmann , Mouad Edderkaoui , Adrian Lim , Xu Zhou , Katrin Pfütze , Angela Schulz , Stephan Wolf , Kai Hu , Christoph Springfeld , Sadaf S. Mughal , Lenart Zezlina , Franco Fortunato , Georg Beyer , Julia Mayerle , Susanne Roth , Johannes Hulkkonen , Daniela Merz , John P. Neoptolemos
Background
Despite some recent advances, pancreatic ductal adenocarcinoma (PDAC) remains a growing oncological challenge. New drugs capable of targeting more than one oncogenic pathway may be one way to improve patient outcomes. This study characterizes the effectiveness of Metavert a first-in-class dual inhibitor of GSK3-β and histone deacetylase in treating PDAC as a single agent or in combination with standard cytotoxics.
Methods
Thirty-six Patient-Derived Organoids (hPDOs) characterised by RNASeq and whole exome sequencing were treated with Metavert alone or in combination with standard cytotoxics. Transcriptomic signatures (TS) representing sensitivity to Metavert alone or sensitivity to Metavert + irinotecan (IR) were evaluated in 47 patient samples, chemo-naïve in 26 and post-chemotherapy in 21 (gemcitabine=5; FOLFIRINOX=14, both=2) with companion multiplexed immunofluorescence and RNASeq data.
Results
Metavert combined with gemcitabine, irinotecan, 5FU, oxaliplatin, and paclitaxel was synergistic in the hPDOs. Basal-subtype hPDOs were more sensitive to Metavert alone whereas the Metavert+IR combination exhibited synergy in Classical-subtype hPDOs with increased apoptosis and autophagy. hPDO-derived TS evaluated in PDAC tissues demonstrated that Metavert-TSHi samples were enriched for mRNA splicing and DNA repair processes; they were associated with Basal-like tissues but also with GATA6+ve-chemo-naïve samples and were higher following gemcitabine but not FOLFIRINOX treatment. In contrast, Metavert+IR-TSHI samples were enriched for TP53 pathways; they were associated with Classical-like pretreatment samples and with GATA6+ve/KRT17+ve hybrid cell types following FOLFIRINOX, but not gemcitabine treatment, and were unrelated to transcriptional subtypes.
Conclusions
Metavert as a single agent and in combination with irinotecan offers novel strategies for treating pancreatic cancer.
{"title":"Metavert synergises with standard cytotoxics in human PDAC organoids and is associated with transcriptomic signatures of therapeutic response","authors":"Jingyu An , Roma Kurilov , Teresa Peccerella , Frank Bergmann , Mouad Edderkaoui , Adrian Lim , Xu Zhou , Katrin Pfütze , Angela Schulz , Stephan Wolf , Kai Hu , Christoph Springfeld , Sadaf S. Mughal , Lenart Zezlina , Franco Fortunato , Georg Beyer , Julia Mayerle , Susanne Roth , Johannes Hulkkonen , Daniela Merz , John P. Neoptolemos","doi":"10.1016/j.tranon.2024.102109","DOIUrl":"10.1016/j.tranon.2024.102109","url":null,"abstract":"<div><h3>Background</h3><p>Despite some recent advances, pancreatic ductal adenocarcinoma (PDAC) remains a growing oncological challenge. New drugs capable of targeting more than one oncogenic pathway may be one way to improve patient outcomes. This study characterizes the effectiveness of Metavert a first-in-class dual inhibitor of GSK3-β and histone deacetylase in treating PDAC as a single agent or in combination with standard cytotoxics.</p></div><div><h3>Methods</h3><p>Thirty-six Patient-Derived Organoids (hPDOs) characterised by RNASeq and whole exome sequencing were treated with Metavert alone or in combination with standard cytotoxics. Transcriptomic signatures (TS) representing sensitivity to Metavert alone or sensitivity to Metavert + irinotecan (IR) were evaluated in 47 patient samples, chemo-naïve in 26 and post-chemotherapy in 21 (gemcitabine=5; FOLFIRINOX=14, both=2) with companion multiplexed immunofluorescence and RNASeq data.</p></div><div><h3>Results</h3><p>Metavert combined with gemcitabine, irinotecan, 5FU, oxaliplatin, and paclitaxel was synergistic in the hPDOs. Basal-subtype hPDOs were more sensitive to Metavert alone whereas the Metavert+IR combination exhibited synergy in Classical-subtype hPDOs with increased apoptosis and autophagy. hPDO-derived TS evaluated in PDAC tissues demonstrated that Metavert-TS<sup>Hi</sup> samples were enriched for mRNA splicing and DNA repair processes; they were associated with Basal-like tissues but also with GATA6<sup>+ve</sup>-chemo-naïve samples and were higher following gemcitabine but not FOLFIRINOX treatment. In contrast, Metavert+IR-TS<sup>HI</sup> samples were enriched for TP53 pathways; they were associated with Classical-like pretreatment samples and with GATA6<sup>+ve</sup>/KRT17<sup>+ve</sup> hybrid cell types following FOLFIRINOX, but not gemcitabine treatment, and were unrelated to transcriptional subtypes.</p></div><div><h3>Conclusions</h3><p>Metavert as a single agent and in combination with irinotecan offers novel strategies for treating pancreatic cancer.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102109"},"PeriodicalIF":5.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002365/pdfft?md5=f85533617deafcbac6264f8bce5bbb23&pid=1-s2.0-S1936523324002365-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142095503","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-31DOI: 10.1016/j.tranon.2024.102093
Xin Chen , Dan Zhang , Haibin Ou , Jing Su , You Wang , Fuxiang Zhou
Background
This study aims to identify key glycosyltransferases (GTs) in colorectal cancer (CRC) and establish a robust prognostic signature derived from GTs.
Methods
Utilizing the AUCell, UCell, singscore, ssgsea, and AddModuleScore algorithms, along with correlation analysis, we redefined genes related to GTs in CRC at the single-cell RNA level. To improve risk model accuracy, univariate Cox and lasso regression were employed to discover a more clinically subset of GTs in CRC. Subsequently, the efficacy of seven machine learning algorithms for CRC prognosis was assessed, focusing on survival outcomes through nested cross-validation. The model was then validated across four independent external cohorts, exploring variations in the tumor microenvironment (TME), response to immunotherapy, mutational profiles, and pathways of each risk group. Importantly, we identified potential therapeutic agents targeting patients categorized into the high-GARS group.
Results
In our research, we classified CRC patients into distinct subgroups, each exhibiting variations in prognosis, clinical characteristics, pathway enrichments, immune infiltration, and immune checkpoint genes expression. Additionally, we established a Glycosyltransferase-Associated Risk Signature (GARS) based on machine learning. GARS surpasses traditional clinicopathological features in both prognostic power and survival prediction accuracy, and it correlates with higher malignancy levels, providing valuable insights into CRC patients. Furthermore, we explored the association between the risk score and the efficacy of immunotherapy.
Conclusion
A prognostic model based on GTs was developed to forecast the response to immunotherapy, offering a novel approach to CRC management.
{"title":"Bulk and single-cell RNA sequencing analyses coupled with multiple machine learning to develop a glycosyltransferase associated signature in colorectal cancer","authors":"Xin Chen , Dan Zhang , Haibin Ou , Jing Su , You Wang , Fuxiang Zhou","doi":"10.1016/j.tranon.2024.102093","DOIUrl":"10.1016/j.tranon.2024.102093","url":null,"abstract":"<div><h3>Background</h3><p>This study aims to identify key glycosyltransferases (GTs) in colorectal cancer (CRC) and establish a robust prognostic signature derived from GTs.</p></div><div><h3>Methods</h3><p>Utilizing the AUCell, UCell, singscore, ssgsea, and AddModuleScore algorithms, along with correlation analysis, we redefined genes related to GTs in CRC at the single-cell RNA level. To improve risk model accuracy, univariate Cox and lasso regression were employed to discover a more clinically subset of GTs in CRC. Subsequently, the efficacy of seven machine learning algorithms for CRC prognosis was assessed, focusing on survival outcomes through nested cross-validation. The model was then validated across four independent external cohorts, exploring variations in the tumor microenvironment (TME), response to immunotherapy, mutational profiles, and pathways of each risk group. Importantly, we identified potential therapeutic agents targeting patients categorized into the high-GARS group.</p></div><div><h3>Results</h3><p>In our research, we classified CRC patients into distinct subgroups, each exhibiting variations in prognosis, clinical characteristics, pathway enrichments, immune infiltration, and immune checkpoint genes expression. Additionally, we established a Glycosyltransferase-Associated Risk Signature (GARS) based on machine learning. GARS surpasses traditional clinicopathological features in both prognostic power and survival prediction accuracy, and it correlates with higher malignancy levels, providing valuable insights into CRC patients. Furthermore, we explored the association between the risk score and the efficacy of immunotherapy.</p></div><div><h3>Conclusion</h3><p>A prognostic model based on GTs was developed to forecast the response to immunotherapy, offering a novel approach to CRC management.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102093"},"PeriodicalIF":5.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002201/pdfft?md5=9416db9a299d204e6a87e7324cbb1a5f&pid=1-s2.0-S1936523324002201-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142096207","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}