首页 > 最新文献

Translational Oncology最新文献

英文 中文
Tumor perfusion enhancement by focus ultrasound-induced blood-brain barrier opening to potentiate anti-PD-1 immunotherapy of glioma 聚焦超声诱导的血脑屏障开放增强肿瘤灌注,为胶质瘤的抗PD-1免疫疗法增效
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-31 DOI: 10.1016/j.tranon.2024.102115
Haiyan Shan , Guangrong Zheng , Shasha Bao , Haiyan Yang , Ujen Duwal Shrestha , Guochen Li , Xirui Duan , Xiaolan Du , Tengfei Ke , Chengde Liao

Objective

To demonstrate the feasibility of using focused ultrasound to enhance delivery of PD-1 inhibitors in glioma rats and determine if such an approach increases treatment efficacy.

Methods

C6 glioma in situ rat model was used in this study. Transcranial irradiation with FUS combined with microbubbles was administered to open the blood-brain barrier (BBB). The efficacy of BBB opening was evaluated in normal rats. The rats with glioma were grouped to evaluate the role of PD-1 inhibitors combined with FUS-induced immune responses in suppressing glioma when the BBB opens. Flow cytometry was used to examine the changes of immune cell populations of lymphocytes in peripheral blood, tumor tissue and spleen tissue of the rats. A section of rat brain tissue was also used for histological and immunohistochemical analysis. The survival of the rats was then monitored; the tumor progression and changes in blood perfusion of tumor were dynamically observed in vivo using multimodal MRI.

Results

FUS combined with microbubbles could enhance the blood perfusion of tumors by increasing the permeability of BBB (p < 0.0001), thus promoting the infiltration of CD4+ T lymphocytes (p < 0.01). Compared with the control group, the combination treatment group had increased in the infiltration number of CD4+(p < 0.05) and CD8+ T (p < 0.05); the tumor volume of the combined treatment group was smaller than that of the control group (p < 0.01) and the survival rate of the rats was prolonged (p < 0.05).

Conclusions

In this study, we demonstrated that the transient opening of the BBB induced by FUS enhanced tumor vascular perfusion and facilitated the delivery of PD-1 inhibitors, ultimately improving the therapeutic efficacy for glioblastoma.

目的 证明在胶质瘤大鼠中使用聚焦超声增强 PD-1 抑制剂递送的可行性,并确定这种方法是否能提高疗效。本研究采用 C6 脑胶质瘤原位大鼠模型,用 FUS 结合微气泡进行经颅照射,以打开血脑屏障(BBB)。在正常大鼠身上评估了打开 BBB 的疗效。将患有胶质瘤的大鼠分组,以评估 PD-1 抑制剂结合 FUS 诱导的免疫反应在 BBB 打开时抑制胶质瘤的作用。流式细胞术用于检测大鼠外周血、肿瘤组织和脾脏组织中淋巴细胞等免疫细胞群的变化。大鼠脑组织切片也被用于组织学和免疫组化分析。结果 FUS 联合微气泡可通过增加 BBB 的通透性来增强肿瘤的血液灌注(p <0.0001),从而促进 CD4+ T 淋巴细胞的浸润(p <0.01)。与对照组相比,联合治疗组的CD4+(p < 0.05)和CD8+ T(p < 0.05)浸润数量增加;联合治疗组的肿瘤体积小于对照组(p < 0.结论在这项研究中,我们证明了FUS诱导的BBB瞬时开放增强了肿瘤血管灌注,促进了PD-1抑制剂的递送,最终提高了胶质母细胞瘤的疗效。
{"title":"Tumor perfusion enhancement by focus ultrasound-induced blood-brain barrier opening to potentiate anti-PD-1 immunotherapy of glioma","authors":"Haiyan Shan ,&nbsp;Guangrong Zheng ,&nbsp;Shasha Bao ,&nbsp;Haiyan Yang ,&nbsp;Ujen Duwal Shrestha ,&nbsp;Guochen Li ,&nbsp;Xirui Duan ,&nbsp;Xiaolan Du ,&nbsp;Tengfei Ke ,&nbsp;Chengde Liao","doi":"10.1016/j.tranon.2024.102115","DOIUrl":"10.1016/j.tranon.2024.102115","url":null,"abstract":"<div><h3>Objective</h3><p>To demonstrate the feasibility of using focused ultrasound to enhance delivery of PD-1 inhibitors in glioma rats and determine if such an approach increases treatment efficacy.</p></div><div><h3>Methods</h3><p>C6 glioma <em>in situ</em> rat model was used in this study. Transcranial irradiation with FUS combined with microbubbles was administered to open the blood-brain barrier (BBB). The efficacy of BBB opening was evaluated in normal rats. The rats with glioma were grouped to evaluate the role of PD-1 inhibitors combined with FUS-induced immune responses in suppressing glioma when the BBB opens. Flow cytometry was used to examine the changes of immune cell populations of lymphocytes in peripheral blood, tumor tissue and spleen tissue of the rats. A section of rat brain tissue was also used for histological and immunohistochemical analysis. The survival of the rats was then monitored; the tumor progression and changes in blood perfusion of tumor were dynamically observed <em>in vivo</em> using multimodal MRI.</p></div><div><h3>Results</h3><p>FUS combined with microbubbles could enhance the blood perfusion of tumors by increasing the permeability of BBB (<em>p &lt;</em> 0.0001), thus promoting the infiltration of CD4<sup>+</sup> T lymphocytes (<em>p &lt;</em> 0.01). Compared with the control group, the combination treatment group had increased in the infiltration number of CD4<sup>+</sup>(<em>p &lt;</em> 0.05) and CD8<sup>+</sup> T (<em>p &lt;</em> 0.05); the tumor volume of the combined treatment group was smaller than that of the control group (<em>p &lt;</em> 0.01) and the survival rate of the rats was prolonged (<em>p &lt;</em> 0.05).</p></div><div><h3>Conclusions</h3><p>In this study, we demonstrated that the transient opening of the BBB induced by FUS enhanced tumor vascular perfusion and facilitated the delivery of PD-1 inhibitors, ultimately improving the therapeutic efficacy for glioblastoma.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102115"},"PeriodicalIF":5.0,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002420/pdfft?md5=5f84095f56fbaf09ddffe45e6c72753d&pid=1-s2.0-S1936523324002420-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142095504","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}
引用次数: 0
Collagen extracellular matrix promotes gastric cancer immune evasion by activating IL4I1-AHR signaling 胶原细胞外基质通过激活 IL4I1-AHR 信号促进胃癌免疫逃避
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-30 DOI: 10.1016/j.tranon.2024.102113
Xiaowei Zhang , Yang Zhao , Xu Chen

Background

Gastric cancer (GC) remains a significant global health challenge with poor prognosis, partly due to its ability to evade the immune system. The extracellular matrix (ECM), particularly collagen, plays a crucial role in tumor immune evasion, but the underlying mechanisms are not fully understood. This study investigates the role of collagen ECM in promoting immune evasion in gastric cancer by activating the IL4I1-AHR signaling pathway.

Methods

We cultured gastric cancer cells in 3D collagen gels and assessed their immune evasion capabilities by co-culturing with HER2-specific CAR-T cells. The expression of IL4I1 and its metabolites was analyzed, and the role of integrin αvβ1 in mediating the effects of collagen was explored. Additionally, the impact of IL4I1-induced AHR activation on CAR-T cell exhaustion was evaluated, both in vitro and in vivo.

Results

We found that gastric cancer cells cultured on collagen exhibited increased resistance to CAR-T cell cytotoxicity, which was associated with upregulated immune checkpoint molecules and downregulated effector cytokines on CAR-T cells. This was linked to increased IL4I1 expression, which was further induced by integrin αvβ1 signaling within the 3D collagen environment. IL4I1 metabolites, particularly KynA, promoted CAR-T cell exhaustion by activating the AHR pathway, leading to decreased cytotoxicity and tumor growth inhibition.

Conclusions

Our study reveals a novel mechanism by which the collagen ECM facilitates immune evasion in gastric cancer through the activation of IL4I1-AHR signaling, contributing to CAR-T cell exhaustion. Targeting this pathway could potentially enhance the efficacy of CAR-T cell therapy in gastric cancer.

背景胃癌(GC)仍然是全球健康面临的一个重大挑战,其预后较差,部分原因是它能够逃避免疫系统。细胞外基质(ECM),尤其是胶原蛋白,在肿瘤免疫逃避中起着至关重要的作用,但其潜在机制尚未完全明了。本研究探讨了胶原 ECM 通过激活 IL4I1-AHR 信号通路在促进胃癌免疫逃避中的作用。方法我们在三维胶原凝胶中培养胃癌细胞,并通过与 HER2 特异性 CAR-T 细胞共培养评估其免疫逃避能力。我们分析了IL4I1及其代谢产物的表达,并探讨了整合素αvβ1在介导胶原作用中的作用。结果我们发现,在胶原蛋白上培养的胃癌细胞对 CAR-T 细胞的细胞毒性表现出更强的抵抗力,这与上调的免疫检查点分子和下调的 CAR-T 细胞效应细胞因子有关。这与三维胶原环境中整合素αvβ1信号进一步诱导的IL4I1表达增加有关。IL4I1 代谢物,尤其是 KynA,通过激活 AHR 通路促进 CAR-T 细胞衰竭,导致细胞毒性降低和肿瘤生长抑制。结论我们的研究揭示了一种新的机制,即胶原 ECM 通过激活 IL4I1-AHR 信号促进胃癌的免疫逃避,导致 CAR-T 细胞衰竭。针对这一途径可能会提高 CAR-T 细胞治疗胃癌的疗效。
{"title":"Collagen extracellular matrix promotes gastric cancer immune evasion by activating IL4I1-AHR signaling","authors":"Xiaowei Zhang ,&nbsp;Yang Zhao ,&nbsp;Xu Chen","doi":"10.1016/j.tranon.2024.102113","DOIUrl":"10.1016/j.tranon.2024.102113","url":null,"abstract":"<div><h3>Background</h3><p>Gastric cancer (GC) remains a significant global health challenge with poor prognosis, partly due to its ability to evade the immune system. The extracellular matrix (ECM), particularly collagen, plays a crucial role in tumor immune evasion, but the underlying mechanisms are not fully understood. This study investigates the role of collagen ECM in promoting immune evasion in gastric cancer by activating the IL4I1-AHR signaling pathway.</p></div><div><h3>Methods</h3><p>We cultured gastric cancer cells in 3D collagen gels and assessed their immune evasion capabilities by co-culturing with HER2-specific CAR-T cells. The expression of IL4I1 and its metabolites was analyzed, and the role of integrin αvβ1 in mediating the effects of collagen was explored. Additionally, the impact of IL4I1-induced AHR activation on CAR-T cell exhaustion was evaluated, both <em>in vitro</em> and <em>in vivo</em>.</p></div><div><h3>Results</h3><p>We found that gastric cancer cells cultured on collagen exhibited increased resistance to CAR-T cell cytotoxicity, which was associated with upregulated immune checkpoint molecules and downregulated effector cytokines on CAR-T cells. This was linked to increased IL4I1 expression, which was further induced by integrin αvβ1 signaling within the 3D collagen environment. IL4I1 metabolites, particularly KynA, promoted CAR-T cell exhaustion by activating the AHR pathway, leading to decreased cytotoxicity and tumor growth inhibition.</p></div><div><h3>Conclusions</h3><p>Our study reveals a novel mechanism by which the collagen ECM facilitates immune evasion in gastric cancer through the activation of IL4I1-AHR signaling, contributing to CAR-T cell exhaustion. Targeting this pathway could potentially enhance the efficacy of CAR-T cell therapy in gastric cancer.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102113"},"PeriodicalIF":5.0,"publicationDate":"2024-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002407/pdfft?md5=b2186aa04e3df43ed12146efc5f454bf&pid=1-s2.0-S1936523324002407-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142095502","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}
引用次数: 0
The role of deubiquitinase USP2 in driving bladder cancer progression by stabilizing EZH2 to epigenetically silence SOX1 expression 去泛素化酶 USP2 通过稳定 EZH2 从表观遗传学上抑制 SOX1 的表达,在推动膀胱癌进展中发挥作用
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-27 DOI: 10.1016/j.tranon.2024.102104
Fanghua Xu , Xiangda Xu , Huanhuan Deng , Zhaojun Yu , Jianbiao Huang , Leihong Deng , Haichao Chao

Background

The Ubiquitin-proteasome system (UPS) is known to participate in multiple cellular events. The deubiquitinating enzyme USP2 (ubiquitin-specific protease 2) is involved in the vasculature remodeling process associated with bladder cancer (BLCA). However, the role of USP2 in BLCA progression has not been clearly defined and whether its regulatory mechanism involving EZH2 (Enhancer of Zeste Homolog 2) remains elusive yet.

Methods

Differential expression patterns of USP2 and EZH2 were examined in 46 pairs of BLCA and adjacent normal tissues. USP2 knockdown plasmids were transfected into 5637 and J82 cells to detect its impact on cell proliferation, migration and invasion using CCK-8, EdU, wound healing and transwell assays. The USP2-EZH2-SOX1 cascade was confirmed through Co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation (ChIP) assays. An in vivo verification was conducted using a xenograft model of nude mice.

Results

USP2 was significantly upregulated in BLCA tissues and cells, which was associated with poor clinical prognosis in BLCA patients. USP2 depletion resulted in decreased cell proliferation, migration and invasion in BLCA cells. USP2 stabilized the EZH2 protein by directly binding to it, thereby reducing its ubiquitination. Ectopic introduction of EZH2 restored cell growth and invasion of BLCA cells, which had been inhibited by USP2 silencing. USP2-mediated stabilization of EZH2 promoted the enrichment of histone H3K27me3 and repression of SOX1. Involvement of the USP2-EZH2-SOX1 axis in tumor formation was ultimately verified in vivo.

Conclusion

Our findings reveal that a USP2-EZH2-SOX1 axis orchestrates the interplay between dysregulated USP2 and EZH2-mediated gene epigenetic silencing in BLCA progression.

背景众所周知,泛素-蛋白酶体系统(UPS)参与多种细胞事件。去泛素化酶 USP2(泛素特异性蛋白酶 2)参与了与膀胱癌(BLCA)相关的血管重塑过程。然而,USP2 在膀胱癌进展过程中的作用尚未得到明确定义,其调控机制是否涉及 EZH2(泽斯特同源酶 2)也仍未确定。方法在 46 对膀胱癌和邻近正常组织中检测了 USP2 和 EZH2 的不同表达模式。将 USP2 敲除质粒转染到 5637 和 J82 细胞中,使用 CCK-8、EdU、伤口愈合和透孔试验检测其对细胞增殖、迁移和侵袭的影响。USP2-EZH2-SOX1级联通过共免疫沉淀(Co-IP)和染色质免疫沉淀(ChIP)试验得到了证实。结果USP2在BLCA组织和细胞中显著上调,这与BLCA患者的临床预后不良有关。USP2 缺失会导致 BLCA 细胞的增殖、迁移和侵袭能力下降。USP2 通过直接与 EZH2 蛋白结合来稳定 EZH2 蛋白,从而减少其泛素化。异位引入 EZH2 可恢复 BLCA 细胞的生长和侵袭,而 USP2 沉默则抑制了细胞的生长和侵袭。USP2 介导的 EZH2 稳定化促进了组蛋白 H3K27me3 的富集和 SOX1 的抑制。结论我们的研究结果表明,USP2-EZH2-SOX1 轴协调了 USP2 失调和 EZH2 介导的基因表观遗传沉默在 BLCA 进展中的相互作用。
{"title":"The role of deubiquitinase USP2 in driving bladder cancer progression by stabilizing EZH2 to epigenetically silence SOX1 expression","authors":"Fanghua Xu ,&nbsp;Xiangda Xu ,&nbsp;Huanhuan Deng ,&nbsp;Zhaojun Yu ,&nbsp;Jianbiao Huang ,&nbsp;Leihong Deng ,&nbsp;Haichao Chao","doi":"10.1016/j.tranon.2024.102104","DOIUrl":"10.1016/j.tranon.2024.102104","url":null,"abstract":"<div><h3>Background</h3><p>The Ubiquitin-proteasome system (UPS) is known to participate in multiple cellular events. The deubiquitinating enzyme USP2 (ubiquitin-specific protease 2) is involved in the vasculature remodeling process associated with bladder cancer (BLCA). However, the role of USP2 in BLCA progression has not been clearly defined and whether its regulatory mechanism involving EZH2 (Enhancer of Zeste Homolog 2) remains elusive yet.</p></div><div><h3>Methods</h3><p>Differential expression patterns of USP2 and EZH2 were examined in 46 pairs of BLCA and adjacent normal tissues. USP2 knockdown plasmids were transfected into 5637 and J82 cells to detect its impact on cell proliferation, migration and invasion using CCK-8, EdU, wound healing and transwell assays. The USP2-EZH2-SOX1 cascade was confirmed through Co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation (ChIP) assays. An <em>in vivo</em> verification was conducted using a xenograft model of nude mice.</p></div><div><h3>Results</h3><p>USP2 was significantly upregulated in BLCA tissues and cells, which was associated with poor clinical prognosis in BLCA patients. USP2 depletion resulted in decreased cell proliferation, migration and invasion in BLCA cells. USP2 stabilized the EZH2 protein by directly binding to it, thereby reducing its ubiquitination. Ectopic introduction of EZH2 restored cell growth and invasion of BLCA cells, which had been inhibited by USP2 silencing. USP2-mediated stabilization of EZH2 promoted the enrichment of histone H3K27me3 and repression of SOX1. Involvement of the USP2-EZH2-SOX1 axis in tumor formation was ultimately verified <em>in vivo</em>.</p></div><div><h3>Conclusion</h3><p>Our findings reveal that a USP2-EZH2-SOX1 axis orchestrates the interplay between dysregulated USP2 and EZH2-mediated gene epigenetic silencing in BLCA progression.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102104"},"PeriodicalIF":5.0,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002316/pdfft?md5=ebbfc99a8a5c065a0d6f95c5e88f2de3&pid=1-s2.0-S1936523324002316-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142089701","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}
引用次数: 0
LncRNA LINC00173 inhibits the development of endometrial cancer by interacting with HNRNPC LncRNA LINC00173通过与HNRNPC相互作用抑制子宫内膜癌的发展
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-26 DOI: 10.1016/j.tranon.2024.102105
Zhijuan Zhu , Rong Du , Juan Yu

Background

Previous research has elaborated on the role of long non-coding RNA LINC00173 in the pathogenesis of various cancers; however, our knowledge of its clinical consequences and mechanisms in endometrial cancer (EC) is limited. Our current work is aimed at investigating the effect of LINC00173 in combination with its upstream gene HNRNPC on EC progression.

Methods

LINC00173 and HNRNPC levels were investigated by qRT-PCR or western blotting in EC tissues. The functional roles of HNRNPC and LINC00173 were assessed using transwell, colony formation and CCK-8 assays. A xenograft was used to verify the phenotype of LINC00173 after its overexpression. The regulatory role between HNRNPC and LINC00173 was investigated using RIP and RNA pull-down analysis.

Results

In EC tissues, LINC00173 expression was down-regulated. We observed that increased LINC00173 inhibited EC cell growth and migration. LINC00173 was a downstream target of HNRNPC, and its expression level was elevated by HNRNPC silencing. LINC00173 overexpression shifted part of HNRNPC into the cytoplasm from the nucleus of EC cells. Furthermore, HNRNPC expression was upregulated in EC and its silencing inhibited EC cell malignancy in vitro.

Conclusion

LINC00173 can impair the malignancy of EC cell by interacting with HNRNPC. This finding may contribute to the understanding of the tumorigenic effects of HNRNPC and LINC00173 on EC.

背景以前的研究已经阐述了长非编码RNA LINC00173在多种癌症发病机制中的作用,但我们对其在子宫内膜癌(EC)中的临床后果和机制的了解还很有限。我们目前的工作旨在研究 LINC00173 与其上游基因 HNRNPC 共同作用对子宫内膜癌进展的影响。方法通过 qRT-PCR 或 Western 印迹法检测子宫内膜癌组织中 LINC00173 和 HNRNPC 的水平。通过transwell、集落形成和CCK-8试验评估了HNRNPC和LINC00173的功能作用。异种移植用于验证 LINC00173 过表达后的表型。结果在EC组织中,LINC00173的表达被下调。我们观察到,LINC00173的增加抑制了EC细胞的生长和迁移。LINC00173 是 HNRNPC 的下游靶标,其表达水平在 HNRNPC 沉默后升高。过表达 LINC00173 会使部分 HNRNPC 从 EC 细胞核转移到细胞质中。结论LINC00173可通过与HNRNPC相互作用来损害EC细胞的恶性程度。这一发现可能有助于了解 HNRNPC 和 LINC00173 对 EC 的致瘤作用。
{"title":"LncRNA LINC00173 inhibits the development of endometrial cancer by interacting with HNRNPC","authors":"Zhijuan Zhu ,&nbsp;Rong Du ,&nbsp;Juan Yu","doi":"10.1016/j.tranon.2024.102105","DOIUrl":"10.1016/j.tranon.2024.102105","url":null,"abstract":"<div><h3>Background</h3><p>Previous research has elaborated on the role of long non-coding RNA LINC00173 in the pathogenesis of various cancers; however, our knowledge of its clinical consequences and mechanisms in endometrial cancer (EC) is limited. Our current work is aimed at investigating the effect of LINC00173 in combination with its upstream gene HNRNPC on EC progression.</p></div><div><h3>Methods</h3><p>LINC00173 and HNRNPC levels were investigated by qRT-PCR or western blotting in EC tissues. The functional roles of HNRNPC and LINC00173 were assessed using transwell, colony formation and CCK-8 assays. A xenograft was used to verify the phenotype of LINC00173 after its overexpression. The regulatory role between HNRNPC and LINC00173 was investigated using RIP and RNA pull-down analysis.</p></div><div><h3>Results</h3><p>In EC tissues, LINC00173 expression was down-regulated. We observed that increased LINC00173 inhibited EC cell growth and migration. LINC00173 was a downstream target of HNRNPC, and its expression level was elevated by HNRNPC silencing. LINC00173 overexpression shifted part of HNRNPC into the cytoplasm from the nucleus of EC cells. Furthermore, HNRNPC expression was upregulated in EC and its silencing inhibited EC cell malignancy <em>in vitro</em>.</p></div><div><h3>Conclusion</h3><p>LINC00173 can impair the malignancy of EC cell by interacting with HNRNPC. This finding may contribute to the understanding of the tumorigenic effects of HNRNPC and LINC00173 on EC.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102105"},"PeriodicalIF":5.0,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002328/pdfft?md5=14dc400e2bd75aba64ce82f6b23b2b15&pid=1-s2.0-S1936523324002328-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142076772","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}
引用次数: 0
Neratinib plus dasatinib is highly synergistic in HER2-positive breast cancer in vitro and in vivo 奈拉替尼加达沙替尼在体外和体内对HER2阳性乳腺癌具有高度协同作用
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-26 DOI: 10.1016/j.tranon.2024.102073
Neil T Conlon , Sandra Roche , Amira F Mahdi , Alacoque Browne , Laura Breen , Johanna Gaubatz , Justine Meiller , Fiona O'Neill , Lorraine O'Driscoll , Mattia Cremona , Bryan T Hennessy , Lisa D Eli , John Crown , Denis M Collins

Background

HER2-targeted therapies have revolutionised the treatment of HER2-positive breast cancer. However, de novo resistance or the emergence of acquired resistance is a persistent clinical problem. Here we report that neratinib, an irreversible pan-HER inhibitor, in combination with the multi-kinase inhibitor dasatinib, currently used to treat certain leukemias, has strong anti-proliferative effects against models of HER2-positive breast cancer that are innately resistant to trastuzumab or have acquired resistance to neratinib.

Methods

Neratinib plus dasatinib was examined in a panel of 20 breast cancer cell lines, including HER2-positive, estrogen-receptor-positive, triple negative, and acquired HER2-targeted therapy resistant models. Drug effects on migration and apoptosis induction was evaluated and signaling alterations were determined by reverse phase protein array (RPPA). In vivo efficacy was examined using orthotopically-implanted HCC1954 cells.

Results

Synergy was observed in cell lines innately resistant to trastuzumab, models with acquired resistance to neratinib, and in triple negative breast cancer cell lines. Further investigation showed that neratinib plus dasatinib induced apoptosis and inhibited cell migration to a greater degree than either drug alone. RPPA revealed that the combination caused suppression of key survival signaling through EGFR, Akt, and MAPK inhibition. In vivo, neratinib plus dasatinib was well tolerated and had a prolonged anti-tumor effect against HCC1954 xenografts.

Conclusions

This study provides a strong pre-clinical rationale for the clinical investigation neratinib and dasatinib in HER2+ breast cancer.

背景HER2靶向疗法彻底改变了HER2阳性乳腺癌的治疗。然而,新发耐药性或获得性耐药性的出现是一个长期存在的临床问题。我们在此报告,奈拉替尼是一种不可逆的泛HER抑制剂,与目前用于治疗某些白血病的多激酶抑制剂达沙替尼联用,对曲妥珠单抗先天耐药或奈拉替尼获得性耐药的HER2阳性乳腺癌模型具有很强的抗增殖作用。方法 在20种乳腺癌细胞系(包括HER2阳性、雌激素受体阳性、三阴性和获得性HER2靶向治疗耐药模型)中对奈拉替尼加达沙替尼进行了研究。评估了药物对迁移和凋亡诱导的影响,并通过反相蛋白阵列(RPPA)确定了信号的改变。结果在对曲妥珠单抗先天耐药的细胞系、对奈拉替尼获得性耐药的模型以及三阴性乳腺癌细胞系中观察到了协同作用。进一步研究表明,与单独使用两种药物相比,奈拉替尼加达沙替尼诱导细胞凋亡和抑制细胞迁移的作用更强。RPPA显示,联合用药通过抑制表皮生长因子受体、Akt和MAPK,抑制了关键的生存信号传导。在体内,奈拉替尼加达沙替尼的耐受性良好,对HCC1954异种移植物的抗肿瘤作用持续时间长。
{"title":"Neratinib plus dasatinib is highly synergistic in HER2-positive breast cancer in vitro and in vivo","authors":"Neil T Conlon ,&nbsp;Sandra Roche ,&nbsp;Amira F Mahdi ,&nbsp;Alacoque Browne ,&nbsp;Laura Breen ,&nbsp;Johanna Gaubatz ,&nbsp;Justine Meiller ,&nbsp;Fiona O'Neill ,&nbsp;Lorraine O'Driscoll ,&nbsp;Mattia Cremona ,&nbsp;Bryan T Hennessy ,&nbsp;Lisa D Eli ,&nbsp;John Crown ,&nbsp;Denis M Collins","doi":"10.1016/j.tranon.2024.102073","DOIUrl":"10.1016/j.tranon.2024.102073","url":null,"abstract":"<div><h3>Background</h3><p>HER2-targeted therapies have revolutionised the treatment of HER2-positive breast cancer. However, <em>de novo</em> resistance or the emergence of acquired resistance is a persistent clinical problem. Here we report that neratinib, an irreversible pan-HER inhibitor, in combination with the multi-kinase inhibitor dasatinib, currently used to treat certain leukemias, has strong anti-proliferative effects against models of HER2-positive breast cancer that are innately resistant to trastuzumab or have acquired resistance to neratinib.</p></div><div><h3>Methods</h3><p>Neratinib plus dasatinib was examined in a panel of 20 breast cancer cell lines, including HER2-positive, estrogen-receptor-positive, triple negative, and acquired HER2-targeted therapy resistant models. Drug effects on migration and apoptosis induction was evaluated and signaling alterations were determined by reverse phase protein array (RPPA). <em>In vivo</em> efficacy was examined using orthotopically-implanted HCC1954 cells.</p></div><div><h3>Results</h3><p>Synergy was observed in cell lines innately resistant to trastuzumab, models with acquired resistance to neratinib, and in triple negative breast cancer cell lines. Further investigation showed that neratinib plus dasatinib induced apoptosis and inhibited cell migration to a greater degree than either drug alone. RPPA revealed that the combination caused suppression of key survival signaling through EGFR, Akt, and MAPK inhibition. <em>In vivo</em>, neratinib plus dasatinib was well tolerated and had a prolonged anti-tumor effect against HCC1954 xenografts.</p></div><div><h3>Conclusions</h3><p>This study provides a strong pre-clinical rationale for the clinical investigation neratinib and dasatinib in HER2+ breast cancer.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102073"},"PeriodicalIF":5.0,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002006/pdfft?md5=8714f7eb08b0c65d849078a50d99b7be&pid=1-s2.0-S1936523324002006-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142076774","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}
引用次数: 0
Therapeutic potential of tLyp-1-EV-shCTCF in inhibiting liver cancer stem cell self-renewal and immune escape via SALL3 modulation in hepatocellular carcinoma tLyp-1-EV-shCTCF 通过调节 SALL3 抑制肝癌干细胞自我更新和免疫逃逸的治疗潜力
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-25 DOI: 10.1016/j.tranon.2024.102048
Heng Zhu , Zhihui Xie

The progression of hepatocellular carcinoma (HCC) is influenced by disrupted metabolic processes, presenting challenges in prognostic outcomes. Hepatocellular carcinoma (HCC), a leading cause of cancer-related mortality, is closely associated with metabolic reprogramming and stem cell-like properties in liver cancer stem cells (LCSCs). This study explored the potential molecular mechanisms by which tLyP-1-modified extracellular vesicles (EVs) delivering CTCF shRNA (tLyp-1-EV-shCTCF) regulate mitochondrial DNA methylation-induced glycolytic metabolic reprogramming and LCSC self-renewal. Through a series of methods, including Western blot, nanoparticle tracking analysis, and immunofluorescence, we demonstrated the successful delivery and internalization of tLyp-1-EV in HCC cells. Our results identified SALL3 as a critical factor underexpressed in HCC and LCSCs, while CTCF was overexpressed. Overexpression of SALL3 inhibited LCSC self-renewal and immune evasion by blocking the CTCF-DNMT3A interaction, thus repressing DNMT3A methyltransferase activity and subsequent mitochondrial DNA methylation-mediated glycolytic metabolic reprogramming. In vivo experiments further supported these findings, showing that tLyp-1-EV-shCTCF treatment significantly reduced tumor growth by upregulating SALL3 expression, thereby inhibiting glycolytic metabolic reprogramming and enhancing the immune response against HCC cells. This study provides novel insights into the role of SALL3 and mitochondrial DNA methylation in HCC progression, offering potential therapeutic targets for combating HCC and its stem cell-like properties.

肝细胞癌(HCC)的进展受代谢过程紊乱的影响,给预后结果带来了挑战。肝细胞癌(HCC)是癌症相关死亡的主要原因,它与肝癌干细胞(LCSCs)的代谢重编程和干细胞样特性密切相关。本研究探索了tLyP-1修饰的细胞外囊泡递送CTCF shRNA(tLyp-1-EV-shCTCF)调控线粒体DNA甲基化诱导的糖酵解代谢重编程和LCSC自我更新的潜在分子机制。通过Western印迹、纳米颗粒追踪分析和免疫荧光等一系列方法,我们证明了tLyp-1-EV在HCC细胞中的成功递送和内化。我们的研究结果发现,SALL3 是 HCC 和 LCSCs 中表达不足的关键因子,而 CTCF 则表达过高。SALL3 的过表达通过阻断 CTCF-DNMT3A 的相互作用抑制了 LCSC 的自我更新和免疫逃避,从而抑制了 DNMT3A 甲基转移酶的活性以及随后线粒体 DNA 甲基化介导的糖酵解代谢重编程。体内实验进一步支持了这些发现,实验结果表明,tLyp-1-EV-shCTCF 治疗通过上调 SALL3 的表达,从而抑制了糖酵解代谢重编程,增强了针对 HCC 细胞的免疫反应,从而显著降低了肿瘤的生长。这项研究为SALL3和线粒体DNA甲基化在HCC进展中的作用提供了新的见解,为抗击HCC及其干细胞样特性提供了潜在的治疗靶点。
{"title":"Therapeutic potential of tLyp-1-EV-shCTCF in inhibiting liver cancer stem cell self-renewal and immune escape via SALL3 modulation in hepatocellular carcinoma","authors":"Heng Zhu ,&nbsp;Zhihui Xie","doi":"10.1016/j.tranon.2024.102048","DOIUrl":"10.1016/j.tranon.2024.102048","url":null,"abstract":"<div><p>The progression of hepatocellular carcinoma (HCC) is influenced by disrupted metabolic processes, presenting challenges in prognostic outcomes. Hepatocellular carcinoma (HCC), a leading cause of cancer-related mortality, is closely associated with metabolic reprogramming and stem cell-like properties in liver cancer stem cells (LCSCs). This study explored the potential molecular mechanisms by which tLyP-1-modified extracellular vesicles (EVs) delivering CTCF shRNA (tLyp-1-EV-shCTCF) regulate mitochondrial DNA methylation-induced glycolytic metabolic reprogramming and LCSC self-renewal. Through a series of methods, including Western blot, nanoparticle tracking analysis, and immunofluorescence, we demonstrated the successful delivery and internalization of tLyp-1-EV in HCC cells. Our results identified SALL3 as a critical factor underexpressed in HCC and LCSCs, while CTCF was overexpressed. Overexpression of SALL3 inhibited LCSC self-renewal and immune evasion by blocking the CTCF-DNMT3A interaction, thus repressing DNMT3A methyltransferase activity and subsequent mitochondrial DNA methylation-mediated glycolytic metabolic reprogramming. <em>In vivo</em> experiments further supported these findings, showing that tLyp-1-EV-shCTCF treatment significantly reduced tumor growth by upregulating SALL3 expression, thereby inhibiting glycolytic metabolic reprogramming and enhancing the immune response against HCC cells. This study provides novel insights into the role of SALL3 and mitochondrial DNA methylation in HCC progression, offering potential therapeutic targets for combating HCC and its stem cell-like properties.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102048"},"PeriodicalIF":5.0,"publicationDate":"2024-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S193652332400175X/pdfft?md5=4becfd946383cbafd804d66a255b79fe&pid=1-s2.0-S193652332400175X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142058370","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}
引用次数: 0
The immunological landscape and silico analysis of key paraptosis regulator LPAR1 in gastric cancer patients 胃癌患者体内关键副凋亡调节因子 LPAR1 的免疫学特征和硅学分析
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-24 DOI: 10.1016/j.tranon.2024.102110
Ya-Jie Dai , Hao-Dong Tang , Guang-Qing Jiang , Zhai-Yue Xu

This study aims to identify key regulators of paraptosis in gastric cancer (GC) and explore their potential in guiding therapeutic strategies, especially in stomach adenocarcinoma (STAD). Genes associated with paraptosis were identified from the references and subjected to Cox regression analysis in the TCGA-STAD cohort. Using machine learning models, LPAR1 consistently ranked highest in feature importance. Multiple sequencing data showed that LPAR1 was significantly overexpressed in cancer-associated fibroblasts (CAFs). LPAR1 expression was significantly higher in normal tissues, and ROC analysis demonstrated its discriminative ability. Copy number alterations and microsatellite instability were significantly associated with LPAR1 expression. High LPAR1 expression correlated with advanced tumor grades and specific cancer immune subtypes, and multivariate analysis confirmed LPAR1 as an independent predictor of poor prognosis. LPAR1 expression was associated with different immune response metrics, including immune effector activation and upregulated chemokine secretion. High LPAR1 expression also correlated with increased sensitivity to compounds, such as BET bromodomain inhibitors I-BET151 and RITA, suggesting LPAR1 as a biomarker for predicting drug activity. FOXP2 showed a strong positive correlation with LPAR1 transcriptional regulation, while increased methylation of LPAR1 promoter regions was negatively correlated with gene expression. Knockdown of LPAR1 affected cell growth in most tumor cell lines, and in vitro experiments demonstrated that LPAR1 influenced extracellular matrix (ECM) contraction and cell viability in the paraptosis of CAFs. These findings suggest that LPAR1 is a critical regulator of paraptosis in GC and a potential biomarker for drug sensitivity and immunotherapy response. This underscores the role of CAFs in mediating tumorigenic effects and suggests that targeting LPAR1 could be a promising strategy for precision medicine in GC.

本研究旨在确定胃癌(GC)中副aptosis的关键调控因子,并探索它们在指导治疗策略方面的潜力,尤其是在胃腺癌(STAD)中。从参考文献中确定了与副aptosis相关的基因,并在TCGA-STAD队列中对其进行了Cox回归分析。通过机器学习模型,LPAR1的特征重要性一直名列前茅。多重测序数据显示,LPAR1在癌症相关成纤维细胞(CAFs)中显著过表达。LPAR1在正常组织中的表达明显较高,ROC分析表明了它的鉴别能力。拷贝数改变和微卫星不稳定性与LPAR1的表达明显相关。LPAR1的高表达与晚期肿瘤分级和特定癌症免疫亚型相关,多变量分析证实LPAR1是不良预后的独立预测因子。LPAR1的表达与不同的免疫反应指标相关,包括免疫效应因子激活和趋化因子分泌上调。LPAR1的高表达还与对化合物(如BET溴域抑制剂I-BET151和RITA)的敏感性增加有关,这表明LPAR1是预测药物活性的生物标记物。FOXP2与LPAR1的转录调控呈强正相关,而LPAR1启动子区域甲基化的增加与基因表达呈负相关。敲除 LPAR1 会影响大多数肿瘤细胞系的细胞生长,体外实验表明,LPAR1 会影响细胞外基质(ECM)的收缩和 CAFs 副凋亡过程中的细胞活力。这些研究结果表明,LPAR1 是 GC 副aptosis 的关键调节因子,也是药物敏感性和免疫疗法反应的潜在生物标志物。这强调了CAFs在介导致瘤效应中的作用,并表明以LPAR1为靶点可能是GC精准医疗的一种有前途的策略。
{"title":"The immunological landscape and silico analysis of key paraptosis regulator LPAR1 in gastric cancer patients","authors":"Ya-Jie Dai ,&nbsp;Hao-Dong Tang ,&nbsp;Guang-Qing Jiang ,&nbsp;Zhai-Yue Xu","doi":"10.1016/j.tranon.2024.102110","DOIUrl":"10.1016/j.tranon.2024.102110","url":null,"abstract":"<div><p>This study aims to identify key regulators of paraptosis in gastric cancer (GC) and explore their potential in guiding therapeutic strategies, especially in stomach adenocarcinoma (STAD). Genes associated with paraptosis were identified from the references and subjected to Cox regression analysis in the TCGA-STAD cohort. Using machine learning models, LPAR1 consistently ranked highest in feature importance. Multiple sequencing data showed that LPAR1 was significantly overexpressed in cancer-associated fibroblasts (CAFs). LPAR1 expression was significantly higher in normal tissues, and ROC analysis demonstrated its discriminative ability. Copy number alterations and microsatellite instability were significantly associated with LPAR1 expression. High LPAR1 expression correlated with advanced tumor grades and specific cancer immune subtypes, and multivariate analysis confirmed LPAR1 as an independent predictor of poor prognosis. LPAR1 expression was associated with different immune response metrics, including immune effector activation and upregulated chemokine secretion. High LPAR1 expression also correlated with increased sensitivity to compounds, such as BET bromodomain inhibitors I-BET151 and RITA, suggesting LPAR1 as a biomarker for predicting drug activity. FOXP2 showed a strong positive correlation with LPAR1 transcriptional regulation, while increased methylation of LPAR1 promoter regions was negatively correlated with gene expression. Knockdown of LPAR1 affected cell growth in most tumor cell lines, and in <em>vitro</em> experiments demonstrated that LPAR1 influenced extracellular matrix (ECM) contraction and cell viability in the paraptosis of CAFs. These findings suggest that LPAR1 is a critical regulator of paraptosis in GC and a potential biomarker for drug sensitivity and immunotherapy response. This underscores the role of CAFs in mediating tumorigenic effects and suggests that targeting LPAR1 could be a promising strategy for precision medicine in GC.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102110"},"PeriodicalIF":5.0,"publicationDate":"2024-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002377/pdfft?md5=4b69d1ec237a0405eb8cfb8cdb6240c3&pid=1-s2.0-S1936523324002377-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142049481","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}
引用次数: 0
Transcriptional regulation of DLGAP5 by AR suppresses p53 signaling and inhibits CD8+ T cell infiltration in triple-negative breast cancer AR对DLGAP5的转录调控可抑制p53信号传导并抑制三阴性乳腺癌的CD8+T细胞浸润
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-24 DOI: 10.1016/j.tranon.2024.102081
Qing Pan, Dachang Ma, Yi Xiao, Kun Ji, Jun Wu

Triple-negative breast cancer (TNBC) is a challenging subtype with unclear biological mechanisms. Recently, the transcription factor androgen receptor (AR) and its regulation of the DLGAP5 gene have gained attention in TNBC pathogenesis. In this study, we found a positive correlation between high AR expression and TNBC cell proliferation and growth. Furthermore, we confirmed DLGAP5 as a critical downstream regulator of AR with high expression in TNBC tissues. Knockdown of DLGAP5 significantly inhibited TNBC cell proliferation, migration, and invasion. AR was observed to directly bind to the DLGAP5 promoter, enhancing its transcriptional activity and suppressing the activation of the p53 signaling pathway. In vivo experiments further validated that downregulation of AR or DLGAP5 inhibited tumor growth and enhanced CD8+ T cell infiltration. This study highlights the crucial roles of AR and DLGAP5 in TNBC growth and immune cell infiltration. Taken together, AR inhibits the p53 signaling pathway by promoting DLGAP5 expression, thereby impacting CD8+ T cell infiltration in TNBC.

三阴性乳腺癌(TNBC)是一种具有挑战性的亚型,其生物学机制尚不清楚。最近,转录因子雄激素受体(AR)及其对 DLGAP5 基因的调控在 TNBC 发病机制中受到关注。在这项研究中,我们发现 AR 的高表达与 TNBC 细胞的增殖和生长呈正相关。此外,我们还证实 DLGAP5 是 AR 的一个关键下游调控因子,在 TNBC 组织中高表达。敲除 DLGAP5 能显著抑制 TNBC 细胞的增殖、迁移和侵袭。据观察,AR 可直接与 DLGAP5 启动子结合,增强其转录活性并抑制 p53 信号通路的激活。体内实验进一步验证了下调AR或DLGAP5可抑制肿瘤生长并增强CD8+ T细胞浸润。这项研究强调了AR和DLGAP5在TNBC生长和免疫细胞浸润中的关键作用。综上所述,AR通过促进DLGAP5的表达来抑制p53信号通路,从而影响CD8+ T细胞在TNBC中的浸润。
{"title":"Transcriptional regulation of DLGAP5 by AR suppresses p53 signaling and inhibits CD8+ T cell infiltration in triple-negative breast cancer","authors":"Qing Pan,&nbsp;Dachang Ma,&nbsp;Yi Xiao,&nbsp;Kun Ji,&nbsp;Jun Wu","doi":"10.1016/j.tranon.2024.102081","DOIUrl":"10.1016/j.tranon.2024.102081","url":null,"abstract":"<div><p>Triple-negative breast cancer (TNBC) is a challenging subtype with unclear biological mechanisms. Recently, the transcription factor androgen receptor (AR) and its regulation of the DLGAP5 gene have gained attention in TNBC pathogenesis. In this study, we found a positive correlation between high AR expression and TNBC cell proliferation and growth. Furthermore, we confirmed DLGAP5 as a critical downstream regulator of AR with high expression in TNBC tissues. Knockdown of DLGAP5 significantly inhibited TNBC cell proliferation, migration, and invasion. AR was observed to directly bind to the DLGAP5 promoter, enhancing its transcriptional activity and suppressing the activation of the p53 signaling pathway. <em>In vivo</em> experiments further validated that downregulation of AR or DLGAP5 inhibited tumor growth and enhanced CD8<sup>+</sup> <em>T</em> cell infiltration. This study highlights the crucial roles of AR and DLGAP5 in TNBC growth and immune cell infiltration. Taken together, AR inhibits the p53 signaling pathway by promoting DLGAP5 expression, thereby impacting CD8<sup>+</sup> <em>T</em> cell infiltration in TNBC.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102081"},"PeriodicalIF":5.0,"publicationDate":"2024-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002080/pdfft?md5=0caeea541f8c0ff0adeadb7f7ea2b02a&pid=1-s2.0-S1936523324002080-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142047839","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}
引用次数: 0
Combining Mitomycin C with inhibition of BAD phosphorylation enhances apoptotic cell death in advanced cervical cancer 将丝裂霉素 C 与 BAD 磷酸化抑制剂结合使用,可增强晚期宫颈癌细胞的凋亡能力
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-24 DOI: 10.1016/j.tranon.2024.102103
Liqiong Wang , Xi Zhang , Shu Chen , Qiuhua Ye , Basappa Basappa , Tao Zhu , Peter E. Lobie , Vijay Pandey

Objective

Mitomycin C (MMC), a DNA-damaging chemotherapeutic, is commonly used clinically for recurrent cervical carcinoma (CC), either alone or in combination. MMC generates DNA damage resulting in CC cell death yet also induces increased AKT-BAD phosphorylation associated with drug resistance and reduced clinical benefit. The present study evaluates the efficacy of combined MMC and a BAD phosphorylation inhibitor in CC.

Methods

The association and function of phosphorylation of BAD on serine 99 (pBADS99) for cell survival of both MMC-resistant or sensitive-CC cells was explored. BAD was mutated to BADS99A to examine the requirement of BADS99 for CC cell survival and a novel small-molecule inhibitor of pBADS99 was utilized. Cell proliferation, survival, foci formation, and patient-derived organoids (PDOs) assays were utilized to determine efficacy, synergy and related mechanisms.

Results

MMC IC50 was positively correlated to the cell line pBADS99/BAD ratio. Increased BADS99 phosphorylation was observed in both MMC-sensitive or -resistant CC cells after MMC treatment. Inhibition of pBADS99 in CC cell lines produced synergistic apoptosis through BAD-mediated apoptotic pathways and enhanced DNA damage in response to MMC. The concurrent use of pharmacological inhibition of pBADS99 and MMC was synergistic, resulting in diminished cell viability and inducing apoptotic cell death in MMC-sensitive and -resistant CC cell lines or patient-derived organoids.

Conclusion

A combination of MMC with inhibition of BAD phosphorylation potentiated efficacy compared to single agent treatment. The potential further development of such strategies may provide outcome benefits to patients with CC.

目的:临床上通常单独或联合使用治疗复发性宫颈癌(CC)的脱氧核糖核酸损伤化疗药丝裂霉素 C(MMC)。MMC 会造成 DNA 损伤,导致 CC 细胞死亡,但也会诱导 AKT-BAD 磷酸化增加,从而产生耐药性,降低临床疗效。本研究评估了 MMC 和 BAD 磷酸化抑制剂联合治疗 CC 的疗效。方法探讨了 BAD 在丝氨酸 99 上的磷酸化(pBADS99)与 MMC 耐药或敏感 CC 细胞存活的关联和功能。研究人员将 BAD 突变为 BADS99A,以检测 BADS99 对 CC 细胞存活的要求,并使用了一种新型的 pBADS99 小分子抑制剂。利用细胞增殖、存活、病灶形成和患者衍生器官组织(PDOs)测定来确定疗效、协同作用和相关机制。经 MMC 处理后,在对 MMC 敏感或耐药的 CC 细胞中均观察到 BADS99 磷酸化增加。通过 BAD 介导的凋亡途径抑制 CC 细胞系中的 pBADS99,可产生协同凋亡,并增强对 MMC 的 DNA 损伤反应。在对 MMC 敏感和耐药的 CC 细胞系或患者衍生的器官组织中,同时使用 pBADS99 和 MMC 的药理抑制具有协同作用,可降低细胞活力并诱导细胞凋亡。这种策略的进一步发展可能会为CC患者带来疗效。
{"title":"Combining Mitomycin C with inhibition of BAD phosphorylation enhances apoptotic cell death in advanced cervical cancer","authors":"Liqiong Wang ,&nbsp;Xi Zhang ,&nbsp;Shu Chen ,&nbsp;Qiuhua Ye ,&nbsp;Basappa Basappa ,&nbsp;Tao Zhu ,&nbsp;Peter E. Lobie ,&nbsp;Vijay Pandey","doi":"10.1016/j.tranon.2024.102103","DOIUrl":"10.1016/j.tranon.2024.102103","url":null,"abstract":"<div><h3>Objective</h3><p>Mitomycin C (MMC), a DNA-damaging chemotherapeutic, is commonly used clinically for recurrent cervical carcinoma (CC), either alone or in combination. MMC generates DNA damage resulting in CC cell death yet also induces increased AKT-BAD phosphorylation associated with drug resistance and reduced clinical benefit. The present study evaluates the efficacy of combined MMC and a BAD phosphorylation inhibitor in CC.</p></div><div><h3>Methods</h3><p>The association and function of phosphorylation of BAD on serine 99 (pBADS99) for cell survival of both MMC-resistant or sensitive-CC cells was explored. BAD was mutated to BADS99A to examine the requirement of BADS99 for CC cell survival and a novel small-molecule inhibitor of pBADS99 was utilized. Cell proliferation, survival, foci formation, and patient-derived organoids (PDOs) assays were utilized to determine efficacy, synergy and related mechanisms.</p></div><div><h3>Results</h3><p>MMC IC<sub>50</sub> was positively correlated to the cell line pBADS99/BAD ratio. Increased BADS99 phosphorylation was observed in both MMC-sensitive or -resistant CC cells after MMC treatment. Inhibition of pBADS99 in CC cell lines produced synergistic apoptosis through BAD-mediated apoptotic pathways and enhanced DNA damage in response to MMC. The concurrent use of pharmacological inhibition of pBADS99 and MMC was synergistic, resulting in diminished cell viability and inducing apoptotic cell death in MMC-sensitive and -resistant CC cell lines or patient-derived organoids.</p></div><div><h3>Conclusion</h3><p>A combination of MMC with inhibition of BAD phosphorylation potentiated efficacy compared to single agent treatment. The potential further development of such strategies may provide outcome benefits to patients with CC.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102103"},"PeriodicalIF":5.0,"publicationDate":"2024-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002304/pdfft?md5=f1614516e9ababa5ba2278e41e7bfc03&pid=1-s2.0-S1936523324002304-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142047840","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}
引用次数: 0
Photoimmunotherapy for cancer treatment based on organic small molecules: Recent strategies and future directions 基于有机小分子的癌症治疗光免疫疗法:最新战略和未来方向
IF 5 2区 医学 Q2 Medicine Pub Date : 2024-08-24 DOI: 10.1016/j.tranon.2024.102086
Deming Zhao, Xin Wen, Jiani Wu, Feihong Chen

Photodynamic therapy (PDT) is considered as a promising anticancer approach, owning to its high efficiency and spatiotemporal selectivity. Ample evidence indicated that PDT can trigger immunogenic cell death by releasing antigens that activate immune cells to promote anti-tumor immunity. Nevertheless, the inherent nature of tumors and their complex heterogeneity often limits the efficiency of PDT, which can be overcome with a novel strategy of photo-immunotherapy (PIT) strategy. By exploring the principles of PDT induction and ICD enhancement, combined with other therapies such as chemotherapy or immune checkpoint blockade, the tailored solutions can be designed to address specific challenges of drug resistance, hypoxic conditions, and tumor immunosuppressive microenvironments (TIMEs), which enables targeted enhancement of systemic immunity to address most distant and recurrent cancers. The present article summarizes the specific strategies of PIT and discusses recent existing limitations. More importantly, we anticipate that the perspectives presented herein will help address the clinical translation challenges associated with PIT.

光动力疗法(PDT)因其高效率和时空选择性而被认为是一种前景广阔的抗癌方法。大量证据表明,光动力疗法可通过释放抗原引发免疫性细胞死亡,从而激活免疫细胞,促进抗肿瘤免疫。然而,肿瘤的固有特性及其复杂的异质性往往限制了光动力疗法的效率,而光免疫疗法(PIT)这一新策略可以克服这一问题。通过探索 PDT 诱导和 ICD 增强的原理,并结合化疗或免疫检查点阻断等其他疗法,可以设计出量身定制的解决方案,以应对耐药性、缺氧条件和肿瘤免疫抑制微环境(TIMEs)等特定挑战,从而有针对性地增强全身免疫力,解决大多数远期癌症和复发性癌症的问题。本文总结了 PIT 的具体策略,并讨论了近期存在的局限性。更重要的是,我们希望本文提出的观点将有助于解决与 PIT 相关的临床转化难题。
{"title":"Photoimmunotherapy for cancer treatment based on organic small molecules: Recent strategies and future directions","authors":"Deming Zhao,&nbsp;Xin Wen,&nbsp;Jiani Wu,&nbsp;Feihong Chen","doi":"10.1016/j.tranon.2024.102086","DOIUrl":"10.1016/j.tranon.2024.102086","url":null,"abstract":"<div><p>Photodynamic therapy (PDT) is considered as a promising anticancer approach, owning to its high efficiency and spatiotemporal selectivity. Ample evidence indicated that PDT can trigger immunogenic cell death by releasing antigens that activate immune cells to promote anti-tumor immunity. Nevertheless, the inherent nature of tumors and their complex heterogeneity often limits the efficiency of PDT, which can be overcome with a novel strategy of photo-immunotherapy (PIT) strategy. By exploring the principles of PDT induction and ICD enhancement, combined with other therapies such as chemotherapy or immune checkpoint blockade, the tailored solutions can be designed to address specific challenges of drug resistance, hypoxic conditions, and tumor immunosuppressive microenvironments (TIMEs), which enables targeted enhancement of systemic immunity to address most distant and recurrent cancers. The present article summarizes the specific strategies of PIT and discusses recent existing limitations. More importantly, we anticipate that the perspectives presented herein will help address the clinical translation challenges associated with PIT.</p></div>","PeriodicalId":48975,"journal":{"name":"Translational Oncology","volume":"49 ","pages":"Article 102086"},"PeriodicalIF":5.0,"publicationDate":"2024-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1936523324002134/pdfft?md5=5b1e73366947aecae48c9ea0f362d1a8&pid=1-s2.0-S1936523324002134-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142049438","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}
引用次数: 0
期刊
Translational Oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1