Cancer Biology & TherapyPub Date : 2024-12-31Epub Date: 2024-03-11DOI: 10.1080/15384047.2024.2323765
Xi Jia, Naying Liao, Yunqian Yao, Xutao Guo, Kai Chen, Pengcheng Shi
{"title":"Dynamic evolution of bone marrow adipocyte in B cell acute lymphoblastic leukemia: insights from diagnosis to post-chemotherapy.","authors":"Xi Jia, Naying Liao, Yunqian Yao, Xutao Guo, Kai Chen, Pengcheng Shi","doi":"10.1080/15384047.2024.2323765","DOIUrl":"10.1080/15384047.2024.2323765","url":null,"abstract":"<p><p>Adipocyte is a unique and versatile component of bone marrow microenvironment (BMM). However, the dynamic evolution of Bone Marrow (BM) adipocytes from the diagnosis of B cell Acute Lymphoblastic Leukemia (B-ALL) to the post-treatment state, and how they affect the progression of leukemia, remains inadequately explicated. Primary patient-derived xenograft models (PDXs) and stromal cell co-culture system are employed in this study. We show that the dynamic evolution of BM adipocytes from initial diagnosis of B-ALL to the post-chemotherapy phase, transitioning from cellular depletion in the initial leukemia niche to a fully restored state upon remission. Increased BM adipocytes retards engraftment of B-ALL cells in PDX models and inhibits cells growth of B-ALL in vitro. Mechanistically, the proliferation arrest of B-ALL cells in the context of adipocytes-enrichment niche, might attribute to the presence of adiponectin secreted by adipocytes themselves and the absence of cytokines secreted by mesenchymal stem cell (MSCs). In summary, our findings offer a novel perspective for further in-depth understanding of the dynamic balance between BMM and B-ALL.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2323765"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10936623/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140093445","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2024-12-31Epub Date: 2024-02-15DOI: 10.1080/15384047.2024.2314322
Hongjie Wang, Theo Koob, Jonathan R Fromm, Ajay Gopal, Darrick Carter, André Lieber
{"title":"CD46 and CD59 inhibitors enhance complement-dependent cytotoxicity of anti-CD38 monoclonal antibodies daratumumab and isatuximab in multiple myeloma and other B-cell malignancy cells.","authors":"Hongjie Wang, Theo Koob, Jonathan R Fromm, Ajay Gopal, Darrick Carter, André Lieber","doi":"10.1080/15384047.2024.2314322","DOIUrl":"10.1080/15384047.2024.2314322","url":null,"abstract":"<p><p>Multiple myeloma (MM) is an incurable malignancy of the B-cell lineage. Remarkable progress has been made in the treatment of MM with anti-CD38 monoclonal antibodies such as daratumumab and isatuximab, which can kill MM cells by inducing complement-dependent cytotoxicity (CDC). We showed that the CDC efficacy of daratumumab and isatuximab is limited by membrane complement inhibitors, including CD46 and CD59, which are upregulated in MM cells. We recently developed a small recombinant protein, Ad35K++, which is capable of transiently removing CD46 from the cell surface. We also produced a peptide inhibitor of CD59 (rILYd4). In this study, we tested Ad35K++ and rILYd4 in combination with daratumumab and isatuximab in MM cells as well as in cells from two other B-cell malignancies. We showed that Ad35K++ and rILYd4 increased CDC triggered by daratumumab and isatuximab. The combination of both inhibitors had an additive effect <i>in vitro</i> in primary MM cells as well as <i>in vivo</i> in a mouse xenograft model of MM. Daratumumab and isatuximab treatment of MM lines (without Ad35K++ or rILYd4) resulted in the upregulation of CD46/CD59 and/or survival of CD46<sup>high</sup>/CD59<sup>high</sup> MM cells that escaped the second round of daratumumab and isatuximab treatment. The escape in the second treatment cycle was prevented by the pretreatment of cells with Ad35K++. Overall, our data demonstrate that Ad35K++ and rILYd4 are efficient co-therapeutics of daratumumab and isatuximab, specifically in multi-cycle treatment regimens, and could be used to improve treatment of multiple myeloma.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2314322"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10877974/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139740422","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2024-12-31Epub Date: 2024-05-23DOI: 10.1080/15384047.2024.2355703
Chao Huo, Disheng Wu, Xiaodan Li, Yan Zhang, Baoguang Hu, Taoming Zhang, Jianwei Ren, Tianbao Wang, Yi Liu
{"title":"eIf3a mediates malignant biological behaviors in colorectal cancer through the PI3K/AKT signaling pathway.","authors":"Chao Huo, Disheng Wu, Xiaodan Li, Yan Zhang, Baoguang Hu, Taoming Zhang, Jianwei Ren, Tianbao Wang, Yi Liu","doi":"10.1080/15384047.2024.2355703","DOIUrl":"10.1080/15384047.2024.2355703","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is among the most common gastrointestinal malignancies worldwide. eIF3a is highly expressed in a variety of cancer types, yet its role in CRC remains unclear. We introduced ectopic eIF3a expression in CRC cells to investigate its relevance to various malignant behaviors. Further, we silenced eIF3a to explore its effect on tumor growth in a nude mouse tumor xenograft model. Finally, the molecular mechanisms through which eIF3a regulates malignancy in CRC cells were explored through bioinformatics analysis combined with the use of a specific PI3K inhibitor (LY294002). eIF3a was highly expressed in the peripheral blood and cancer tissue of CRC patients. Malignancy and tumor growth were significantly inhibited by silencing eIF3a, while overexpression promoted malignant behaviors, with a positive correlation between PI3K/AKT activation and eIF3a expression. Taken together, eIF3a plays an oncogenic role in CRC by regulating PI3K/AKT signaling and is a potential biomarker for CRC diagnosis and prognostic monitoring.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2355703"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11123456/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141086889","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2024-12-31Epub Date: 2024-05-30DOI: 10.1080/15384047.2024.2360768
Jing Jiang, Shenghong Shi, Wei Zhang, Chao Li, Long Sun, Qidong Ge, Xujun Li
{"title":"Circ_RPPH1 facilitates progression of breast cancer via miR-1296-5p/TRIM14 axis.","authors":"Jing Jiang, Shenghong Shi, Wei Zhang, Chao Li, Long Sun, Qidong Ge, Xujun Li","doi":"10.1080/15384047.2024.2360768","DOIUrl":"10.1080/15384047.2024.2360768","url":null,"abstract":"<p><p><i>Circular RNA Ribonuclease P</i> <i>RNA Component H1</i> (<i>circ_RPPH1</i>) and microRNA (miRNA) <i>miR-1296-5p</i> play a crucial role in breast cancer (BC), but the molecular mechanism is vague. Evidence showed that <i>miR-1296-5p</i> can activate <i>tripartite motif-containing 14</i> (<i>TRIM14</i>). Clinical indications of eighty BC patients were collected and the <i>circ_RPPH1</i> expression was detected using real-time quantitative PCR. MCF-7 and MDA-MB-231 cells were transfected with overexpression or knockdown of <i>circ_RPPH1</i>, <i>miR-1296-5p</i>, or <i>TRIM14</i>. Cell counting kit-8, cell cloning formation, wound healing, Transwell, and flow cytometry assays were performed to investigate the malignant phenotype of BC. The dual-luciferase reporter gene analyses were applied to reveal the interaction between these target genes. Subcutaneous tumorigenic model mice were established with <i>circ_RPPH1</i> overexpression MDA-MB-231 cells in vivo; the tumor weight and volume, levels of <i>miR-1296-5</i> and <i>TRIM14</i> mRNA were measured. Western blot and immunohistochemistry were used to detect TRIM14 in cells and mice. <i>Circ_RPPH1</i> levels were notably higher in BC patients and have been found to promote cell proliferation, invasion, and migration of BC cells. <i>Circ_RPPH1</i> altered cell cycle and hindered apoptosis. <i>Circ_RPPH1</i> knockdown or <i>miR-1296-5p</i> overexpression inhibited the malignant phenotype of BC. Furthermore, <i>miR-1296-5p</i> knockdown reversed <i>circ_RPPH1</i>'s promotion effects on BC. Interestingly, <i>TRIM14</i> overexpression counteracts the inhibitory effects of <i>miR-1296-5p</i> overexpression and <i>circ_RPPH1</i> silencing on BC. Moreover, in BC tumor-bearing mice, <i>circ_RPPH1</i> overexpression led to increased TRIM14 expression and facilitated tumor growth. <i>Circ_RPPH1</i> enhanced BC progression through <i>miR-1296-5p</i>/<i>TRIM14</i> axis, indicating its potential as a biomarker and therapeutic target in BC.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2360768"},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11141472/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141178606","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2024-12-31Epub Date: 2024-06-12DOI: 10.1080/15384047.2024.2365449
Xuewen Yang, Shisen Li, Chunsheng Xu, Shushang Liu, Xiang Zhang, Bo Lian, Mengbin Li
{"title":"Sirtuin1 (sirt1) regulates the glycolysis pathway and decreases cisplatin chemotherapeutic sensitivity to esophageal squamous cell carcinoma.","authors":"Xuewen Yang, Shisen Li, Chunsheng Xu, Shushang Liu, Xiang Zhang, Bo Lian, Mengbin Li","doi":"10.1080/15384047.2024.2365449","DOIUrl":"10.1080/15384047.2024.2365449","url":null,"abstract":"<p><p>We aimed to evaluate the influence of sirtuin1 (sirt1) on the ESCC chemotherapeutic sensitivity to cisplatin. We used ESCC cell ablation sirt1 for establishing a xenograft mouse tumor model. The tumor volume was then detected. sirt1 was over-expressed significantly in ESCC patients and cells. Moreover, sirt1 knockdown raised ESCC sensitivity to cisplatin. Besides, glycolysis was associated with ESCC cell chemotherapy resistance to cisplatin. Furthermore, sirt1 increased ESCC cells' cisplatin chemosensitivity through HK2. Sirt1 enhanced <i>in vivo</i> ESCC chemosensitivity to cisplatin. Overall, these findings suggested that sirt1 knockdown regulated the glycolysis pathway and raised the ESCC chemotherapeutic sensitivity.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2365449"},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11174053/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141305525","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2024-12-31Epub Date: 2024-07-05DOI: 10.1080/15384047.2024.2373497
Chun Peng, Xiaoqing Li, Yuhui Yao, Yu Nie, Lingyao Fan, Chuandong Zhu
{"title":"MiR-135b-5p promotes cetuximab resistance in colorectal cancer by regulating FOXN3.","authors":"Chun Peng, Xiaoqing Li, Yuhui Yao, Yu Nie, Lingyao Fan, Chuandong Zhu","doi":"10.1080/15384047.2024.2373497","DOIUrl":"10.1080/15384047.2024.2373497","url":null,"abstract":"<p><p>Despite advances in targeted therapies, primary and acquired resistance make the treatment of colorectal cancer (CRC) a pressing issue to be resolved. According to reports, the development of CRC is linked to miRNA dysregulation. Multiple studies have demonstrated that miR-135b-5p has an aberrant expression level between CRC tissues and adjacent tissues. However, it is unclear whether there is a correlation between miR-135b-5p and cetuximab (CTx) resistance in CRC. Use the GEO database to measure miR-135b-5p expression in CRC. Additionally, RT-qPCR was applied to ascertain the production level of miR-135b-5p in three human CRC cells and NCM460 cells. The capacity of cells to migrate and invade was examined utilizing the wound-healing and transwell assays, while the CCK-8 assay served for evaluating cell viability, as well as colony formation assays for proliferation. The expected target protein of miR-135b-5p in CRC cell cetuximab resistance has been investigated using western blot. Suppression of miR-135b-5p could increase the CTx sensitivity of CTx-resistant CRC cells, as manifested by the attenuation of proliferation, migration, and invasion ability. Mechanistic studies revealed miR-135b-5p regulates the epithelial-to-mesenchymal transition (EMT) process and Wnt/β-catenin signaling pathway through downgulating FOXN3. In short, knockdowning miR-135b-5p could increase FOXN3 expression in CRC cells, promote the EMT process, and simultaneously activate the Wnt/β-catenin signaling pathway to elevate CTx resistance in CRC cells.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2373497"},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11229718/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141533711","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Prognostic value of pretreatment procalcitonin and neutrophil-lymphocyte ratio in extensive-stage small-cell lung cancer.","authors":"Dongfang Chen, Jianlin Xu, Yizhuo Zhao, Baohui Han, Runbo Zhong","doi":"10.1080/15384047.2024.2331273","DOIUrl":"10.1080/15384047.2024.2331273","url":null,"abstract":"<p><strong>Background: </strong>To investigate the influence of pretreatment neutrophil-to-lymphocyte ratio (NLR) and procalcitonin (PCT) on progression-free survival (PFS) in extensive-stage small-cell lung cancer (SCLC) patients.</p><p><strong>Method: </strong>A total of 100 extensive-stage SCLC patients were enrolled in our study. Patients were stratified according to the median values of pretreatment NLR and PCT levels: low NLR group (NLR ≤3.17), high NLR group (NLR>3.17), low PCT group (PCT ≤0.06; ng/ml), high PCT group (PCT>0.06; ng/ml). The Kaplan-Meier method and multivariable Cox regression model were used to reveal the prognostic effects of pretreatment NLR and PCT on PFS.</p><p><strong>Results: </strong>The median PFS of the total extensive-stage SCLC patients was 6.0 months. The median PFS of low pretreatment NLR group (NLR ≤3.17) was not significantly different from that of high pretreatment NLR group (6.2 months vs 5.8 months; <i>p</i> = .675). Patients with low pretreatment PCT (PCT ≤0.06; ng/ml) had significantly better PFS than patients with high pretreatment PCT (PCT>0.06; ng/ml) (6.9 months vs 5.7 months; <i>p</i> = .043). With the multivariable Cox regression analysis, the response to first-line chemotherapy (<i>p</i> ≤ .001) and pretreatment PCT (HR = 0.516; 95%CI 0.326-0.817; <i>p</i> = .005) were identified as independent factors associated with PFS.</p><p><strong>Conclusion: </strong>Pretreatment PCT is an independent factor associated with PFS in extensive-stage SCLC patients treated with first-line chemotherapy, but pretreatment NLR reflects no significant prognostic value in our study.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2331273"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10978019/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140304969","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2024-12-31Epub Date: 2024-03-11DOI: 10.1080/15384047.2024.2325130
Jianqing Qiu, Ziyi Zhao, Hongyan Suo, Sarah E Paraghamian, Gabrielle M Hawkins, Wenchuan Sun, Xin Zhang, Tianran Hao, Beor Deng, Xiaochang Shen, Chunxiao Zhou, Victoria Bae-Jump
{"title":"Linoleic acid exhibits anti-proliferative and anti-invasive activities in endometrial cancer cells and a transgenic model of endometrial cancer.","authors":"Jianqing Qiu, Ziyi Zhao, Hongyan Suo, Sarah E Paraghamian, Gabrielle M Hawkins, Wenchuan Sun, Xin Zhang, Tianran Hao, Beor Deng, Xiaochang Shen, Chunxiao Zhou, Victoria Bae-Jump","doi":"10.1080/15384047.2024.2325130","DOIUrl":"10.1080/15384047.2024.2325130","url":null,"abstract":"<p><p>Emerging evidence has provided considerable insights into the integral function of reprogramming fatty acid metabolism in the carcinogenesis and progression of endometrial cancer. Linoleic acid, an essential fatty acid with the highest consumption in the Western diet regimen, has shown pro-tumorigenic or anti-tumorigenic effects on tumor cell growth and invasion in multiple types of cancer. However, the biological role of linoleic acid in endometrial cancer remains unclear. In the present study, we aimed to investigate the functional impact of linoleic acid on cell proliferation, invasion, and tumor growth in endometrial cancer cells and in a transgenic mouse model of endometrial cancer. The results showed that Linoleic acid significantly inhibited the proliferation of endometrial cancer cells in a dose-dependent manner. The treatment of HEC-1A and KLE cells with linoleic acid effectively increased intracellular reactive oxygen species (ROS) production, decreased mitochondrial membrane potential, caused cell cycle G1 arrest, and induced intrinsic and extrinsic apoptosis pathways. The anti-invasive ability of linoleic acid was found to be associated with the epithelial-mesenchymal transition process in both cell lines, including the decreased expression of N-cadherin, snail, and vimentin. Furthermore, treatment of <i>Lkb1</i><sup><i>fl/fl</i></sup><i>p53</i><sup><i>fl/fl</i></sup> transgenic mice with linoleic acid for four weeks significantly reduced the growth of endometrial tumors and decreased the expression of VEGF, vimentin, Ki67, and cyclin D1 in tumor tissues. Our findings demonstrate that linoleic acid exhibits anti-proliferative and anti-invasive activities in endometrial cancer cell lines and the <i>Lkb1</i><sup><i>fl/fl</i></sup><i>p53</i><sup><i>fl/fl</i></sup> mouse model of endometrial cancer, thus providing a pre-clinical basis for future dietary interventions with linoleic acid in endometrial cancer.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2325130"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10936646/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140093446","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2024-12-31Epub Date: 2024-03-14DOI: 10.1080/15384047.2024.2329368
Isil Ezgi Eryilmaz, Unal Egeli, Gulsah Cecener
{"title":"Association between the apoptotic effect of Cabazitaxel and its pro-oxidant efficacy on the redox adaptation mechanisms in prostate cancer cells with different resistance phenotypes.","authors":"Isil Ezgi Eryilmaz, Unal Egeli, Gulsah Cecener","doi":"10.1080/15384047.2024.2329368","DOIUrl":"10.1080/15384047.2024.2329368","url":null,"abstract":"<p><p>Redox adaptation causes poor prognosis by adapting cancer cells to excessive oxidative stress. Previously, we introduced an oxidative stress-resistant metastatic prostate cancer (mPC) model (LNCaP-HPR) that redox adaptation reduced the effect of Cabazitaxel (Cab), the last taxane-derivative for metastatic castration-resistant PC (mCRPC). Whereas, we investigated for the first time whether there is an association between the altered apoptotic effect and pro-oxidant efficacy of Cab on the redox adaptation in PC cells with different phenotypes, including LNCaP mPC, LNCaP-HPR, C4-2 mCRPC, and RWPE-1 cells. Cab was shown pro-oxidant efficacy proportionally with the apoptotic effect, more prominent in the less aggressive LNCaP cells, by increasing the endogenous ROS, mitochondrial damage, and inhibiting nuclear ROS scavengers, p-Nrf2 and HIF-1α. However, the pro-oxidant and apoptotic effect was lower in the LNCaP-HPR and C4-2 cells, indicating that the drug sensitivity of the cells adapted to survive with more ROS was reduced via altered regulation of redox adaptation. Additionally, unlike LNCaP, Cab caused an increase in the p-NF-κB activation, suggesting that the p-NF-κB might accompany maintaining survival with the increased ROS in the aggressive PC cells. Moreover, the cytotoxic and apoptotic effects of Cab were less on RWPE-1 cells compared to LNCaP but were closer to those on the more aggressive LNCaP-HPR and C4-2 cells, except for the changing pro-oxidant effect of Cab. Consequently, this study indicates the variable pro-oxidant effects of Cab on redox-sensitive proteins, which could be a target for improving Cab's apoptotic effect more in aggressive PC cells.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2329368"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10950270/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140130796","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2024-12-31Epub Date: 2024-11-08DOI: 10.1080/15384047.2024.2425127
Shujuan Jin, Mengjiao Zhang, Xiaoting Qiao
{"title":"Cyclophilin A: promising target in cancer therapy.","authors":"Shujuan Jin, Mengjiao Zhang, Xiaoting Qiao","doi":"10.1080/15384047.2024.2425127","DOIUrl":"10.1080/15384047.2024.2425127","url":null,"abstract":"<p><p>Cyclophilin A (CypA), a member of the immunophilin family, stands out as the most prevalent among the cyclophilins found in humans. Beyond serving as the intracellular receptor for the immunosuppressive drug cyclosporine A (CsA), CypA exerts critical functions within the cell via its <i>peptidyl-prolyl cis-trans isomerase</i> (<i>PPIase</i>) activity, which is crucial for processes, such as protein folding, trafficking, assembly, modulation of immune responses, and cell signaling. Increasing evidence indicates that CypA is up-regulated in a variety of human cancers and it may be a novel potential therapeutic target for cancer treatment. Therefore, gaining a thorough understanding of CypA's contribution to cancer could yield fresh perspectives and inform the development of innovative therapeutic approaches. This review delves into the multifaceted roles of CypA in cancer biology and explores the therapeutic potential of targeting CypA.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2425127"},"PeriodicalIF":5.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552246/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142603066","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}