首页 > 最新文献

Cancer research最新文献

英文 中文
Fitness Screens Map State-Specific Glioblastoma Stem Cell Vulnerabilities. 体能筛选绘制出特定状态的胶质母细胞瘤干细胞脆弱性图。
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-02 DOI: 10.1158/0008-5472.CAN-23-4024
Graham MacLeod, Fatemeh Molaei, Shahan Haider, Maira P Almeida, Sichun Lin, Michelle Kushida, Haresh Sureshkumar, Jasmine K Bhatti, Jack Q Lu, Daniel Schramek, Peter B Dirks, Stephane Angers

Glioblastoma (GBM) is the most common and lethal primary brain tumor in adults and is driven by self-renewing glioblastoma stem cells (GSC) that persist after therapy and seed treatment-refractory recurrent tumors. GBM tumors display a high degree of intra- and intertumoral heterogeneity that is a prominent barrier to targeted treatment strategies. This heterogeneity extends to GSCs that exist on a gradient between two transcriptional states or subtypes termed developmental and injury response. Drug targets for each subtype are needed to effectively target GBM. To identify conserved and subtype-specific genetic dependencies across a large and heterogeneous panel of GSCs, we designed the GBM5K-targeted guide RNA library and performed fitness screens in a total of 30 patient-derived GSC cultures. The focused CRISPR screens identified the most conserved subtype-specific vulnerabilities in GSCs and elucidated the functional dependency gradient existing between the developmental and injury response states. Developmental-specific fitness genes were enriched for transcriptional regulators of neurodevelopment, whereas injury response-specific fitness genes were highlighted by several genes implicated in integrin and focal adhesion signaling. These context-specific vulnerabilities conferred differential sensitivity to inhibitors of β1 integrin, focal adhesion kinase, MEK, and OLIG2. Interestingly, the screens revealed that the subtype-specific signaling pathways drive differential cyclin D (CCND1 vs. CCND2) dependencies between subtypes. These data provide a biological insight and mechanistic understanding of GBM heterogeneity and point to opportunities for precision targeting of defined GBM and GSC subtypes to tackle heterogeneity. Significance: CRISPR-Cas9 screens in a panel of patient-derived glioblastoma stem cells reveal heterogeneity in genetic vulnerabilities across subtypes that have important implications for targeted and combination treatment strategies for glioblastoma.

胶质母细胞瘤(GBM)是成人中最常见、最致命的原发性脑肿瘤,由自我更新的胶质母细胞瘤干细胞(GSCs)驱动,这些干细胞在治疗后仍会存活,并为难治性复发肿瘤播下种子。胶质母细胞瘤的瘤内和瘤间具有高度异质性,这是靶向治疗策略的一个突出障碍。这种异质性延伸到 GSC,它们存在于两种转录状态或亚型(称为发育型和损伤反应型)之间的梯度上。要有效针对 GBM,就需要针对每种亚型的药物靶点。为了在大量异质性 GSCs 中鉴定保守的和亚型特异的遗传依赖性,我们设计了 GBM5K 靶向 gRNA 文库,并在总共 30 个患者来源的 GSC 培养物中进行了适配性筛选。聚焦 CRISPR 筛选确定了 GSCs 中最保守的亚型特异性弱点,并阐明了发育和损伤反应状态之间存在的功能依赖梯度。神经发育的转录调控因子富集了发育特异性健壮基因,而损伤反应特异性健壮基因则由几个与整合素和病灶粘附信号转导有关的基因组成。这些特定环境下的脆弱性导致了对β1整合素、FAK、MEK和OLIG2抑制剂的不同敏感性。有趣的是,筛选结果显示,亚型特异性信号通路驱动亚型之间不同的细胞周期蛋白 D(CCND1 与 CCND2)依赖性。这些数据提供了对 GBM 异质性的生物学洞察和机理理解,并为精准靶向确定的 GBM 和 GSC 亚型以解决异质性问题提供了机会。
{"title":"Fitness Screens Map State-Specific Glioblastoma Stem Cell Vulnerabilities.","authors":"Graham MacLeod, Fatemeh Molaei, Shahan Haider, Maira P Almeida, Sichun Lin, Michelle Kushida, Haresh Sureshkumar, Jasmine K Bhatti, Jack Q Lu, Daniel Schramek, Peter B Dirks, Stephane Angers","doi":"10.1158/0008-5472.CAN-23-4024","DOIUrl":"10.1158/0008-5472.CAN-23-4024","url":null,"abstract":"<p><p>Glioblastoma (GBM) is the most common and lethal primary brain tumor in adults and is driven by self-renewing glioblastoma stem cells (GSC) that persist after therapy and seed treatment-refractory recurrent tumors. GBM tumors display a high degree of intra- and intertumoral heterogeneity that is a prominent barrier to targeted treatment strategies. This heterogeneity extends to GSCs that exist on a gradient between two transcriptional states or subtypes termed developmental and injury response. Drug targets for each subtype are needed to effectively target GBM. To identify conserved and subtype-specific genetic dependencies across a large and heterogeneous panel of GSCs, we designed the GBM5K-targeted guide RNA library and performed fitness screens in a total of 30 patient-derived GSC cultures. The focused CRISPR screens identified the most conserved subtype-specific vulnerabilities in GSCs and elucidated the functional dependency gradient existing between the developmental and injury response states. Developmental-specific fitness genes were enriched for transcriptional regulators of neurodevelopment, whereas injury response-specific fitness genes were highlighted by several genes implicated in integrin and focal adhesion signaling. These context-specific vulnerabilities conferred differential sensitivity to inhibitors of β1 integrin, focal adhesion kinase, MEK, and OLIG2. Interestingly, the screens revealed that the subtype-specific signaling pathways drive differential cyclin D (CCND1 vs. CCND2) dependencies between subtypes. These data provide a biological insight and mechanistic understanding of GBM heterogeneity and point to opportunities for precision targeting of defined GBM and GSC subtypes to tackle heterogeneity. Significance: CRISPR-Cas9 screens in a panel of patient-derived glioblastoma stem cells reveal heterogeneity in genetic vulnerabilities across subtypes that have important implications for targeted and combination treatment strategies for glioblastoma.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"3967-3983"},"PeriodicalIF":12.5,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142072097","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Single-Cell Transcriptomic Analysis Identifies Senescent Osteocytes That Trigger Bone Destruction in Breast Cancer Metastasis. 单细胞转录组分析识别出引发乳腺癌转移中骨破坏的衰老骨细胞
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-02 DOI: 10.1158/0008-5472.CAN-24-0857
Japneet Kaur, Manish Adhikari, Hayley M Sabol, Aric Anloague, Sharmin Khan, Noriyoshi Kurihara, Marta Diaz-delCastillo, Christina Møller Andreasen, Charles Lowry Barnes, Jeffrey B Stambough, Michela Palmieri, Olivia Reyes-Castro, Jennifer Zarrer, Hanna Taipaleenmäki, Elena Ambrogini, Maria Almeida, Charles A O'Brien, Intawat Nookaw, Jesus Delgado-Calle

Breast cancer bone metastases increase fracture risk and are a major cause of morbidity and mortality among women. Upon colonization by tumor cells, the bone microenvironment undergoes profound reprogramming to support cancer progression, which disrupts the balance between osteoclasts and osteoblasts and leads to bone lesions. A deeper understanding of the processes mediating this reprogramming could help develop interventions for treating patients with bone metastases. Here, we demonstrated that osteocytes (Ot) in established breast cancer bone metastasis develop premature senescence and a distinctive senescence-associated secretory phenotype (SASP) that favors bone destruction. Single-cell RNA sequencing identified Ots from mice with breast cancer bone metastasis enriched in senescence, SASP markers, and pro-osteoclastogenic genes. Multiplex in situ hybridization and artificial intelligence-assisted analysis depicted Ots with senescence-associated satellite distension, telomere dysfunction, and p16Ink4a expression in mice and patients with breast cancer bone metastasis. Breast cancer cells promoted Ot senescence and enhanced their osteoclastogenic potential in in vitro and ex vivo organ cultures. Clearance of senescent cells with senolytics suppressed bone resorption and preserved bone mass in mice with breast cancer bone metastasis. These results demonstrate that Ots undergo pathological reprogramming by breast cancer cells and identify Ot senescence as an initiating event triggering lytic bone disease in breast cancer metastases. Significance: Breast cancer cells remodel the bone microenvironment by promoting premature cellular senescence and SASP in osteocytes, which can be targeted with senolytics to alleviate bone loss induced by metastatic breast cancer. See related commentary by Frieling and Lynch, p. 3917.

乳腺癌骨转移会增加骨折风险,是妇女发病和死亡的主要原因。肿瘤细胞定植后,骨微环境会发生深刻的重编程,以支持癌症的进展,从而破坏破骨细胞和成骨细胞之间的平衡,导致骨病变。深入了解介导这种重编程的过程有助于开发治疗骨转移患者的干预措施。在这里,我们证明了已确立的乳腺癌骨转移中的骨细胞会出现过早衰老和独特的衰老相关分泌表型(SASP),这种表型有利于骨破坏。单细胞 RNA 测序发现,乳腺癌骨转移小鼠的骨细胞富含衰老、SASP 标记和促破骨细胞生成基因。多重原位杂交和人工智能辅助分析显示,小鼠和乳腺癌骨转移患者的骨细胞存在衰老相关的卫星扩展、端粒功能障碍和 p16Ink4a 表达。乳腺癌细胞促进了骨细胞的衰老,并增强了其在体外和体内器官培养中的破骨细胞生成潜能。用衰老剂清除衰老细胞可抑制骨吸收,保护乳腺癌骨转移小鼠的骨量。这些结果表明,乳腺癌细胞会对成骨细胞进行病理重编程,并确定成骨细胞衰老是引发乳腺癌骨转移中溶解性骨病的起始事件。
{"title":"Single-Cell Transcriptomic Analysis Identifies Senescent Osteocytes That Trigger Bone Destruction in Breast Cancer Metastasis.","authors":"Japneet Kaur, Manish Adhikari, Hayley M Sabol, Aric Anloague, Sharmin Khan, Noriyoshi Kurihara, Marta Diaz-delCastillo, Christina Møller Andreasen, Charles Lowry Barnes, Jeffrey B Stambough, Michela Palmieri, Olivia Reyes-Castro, Jennifer Zarrer, Hanna Taipaleenmäki, Elena Ambrogini, Maria Almeida, Charles A O'Brien, Intawat Nookaw, Jesus Delgado-Calle","doi":"10.1158/0008-5472.CAN-24-0857","DOIUrl":"10.1158/0008-5472.CAN-24-0857","url":null,"abstract":"<p><p>Breast cancer bone metastases increase fracture risk and are a major cause of morbidity and mortality among women. Upon colonization by tumor cells, the bone microenvironment undergoes profound reprogramming to support cancer progression, which disrupts the balance between osteoclasts and osteoblasts and leads to bone lesions. A deeper understanding of the processes mediating this reprogramming could help develop interventions for treating patients with bone metastases. Here, we demonstrated that osteocytes (Ot) in established breast cancer bone metastasis develop premature senescence and a distinctive senescence-associated secretory phenotype (SASP) that favors bone destruction. Single-cell RNA sequencing identified Ots from mice with breast cancer bone metastasis enriched in senescence, SASP markers, and pro-osteoclastogenic genes. Multiplex in situ hybridization and artificial intelligence-assisted analysis depicted Ots with senescence-associated satellite distension, telomere dysfunction, and p16Ink4a expression in mice and patients with breast cancer bone metastasis. Breast cancer cells promoted Ot senescence and enhanced their osteoclastogenic potential in in vitro and ex vivo organ cultures. Clearance of senescent cells with senolytics suppressed bone resorption and preserved bone mass in mice with breast cancer bone metastasis. These results demonstrate that Ots undergo pathological reprogramming by breast cancer cells and identify Ot senescence as an initiating event triggering lytic bone disease in breast cancer metastases. Significance: Breast cancer cells remodel the bone microenvironment by promoting premature cellular senescence and SASP in osteocytes, which can be targeted with senolytics to alleviate bone loss induced by metastatic breast cancer. See related commentary by Frieling and Lynch, p. 3917.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"3936-3952"},"PeriodicalIF":12.5,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11611663/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280503","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Tumor-Associated Microglia Secrete Extracellular ATP to Support Glioblastoma Progression
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-02 DOI: 10.1158/0008-5472.can-24-0018
Caren Yu-Ju Wu, Yiyun Chen, Ya-Jui Lin, Kuo-Chen Wei, Kwang-Yu Chang, Li-Ying Feng, Ko-Ting Chen, Gordon Li, Alexander Liang Ren, Ryan Takeo Nitta, Janet Yuling Wu, Kwang Bog Cho, Ayush Pant, John Choi, Crystal L. Mackall, Lily H. Kim, An-Chih Wu, Jian-Ying Chuang, Chiung-Yin Huang, Christopher M. Jackson, Pin-Yuan Chen, Michael Lim
Glioblastoma (GBM) is a highly aggressive brain tumor with poor prognosis and high recurrence rates. The complex immune microenvironment of GBM is highly infiltrated by tumor-associated microglia and macrophages (TAM). TAMs are known to be heterogeneous in their functional and metabolic states and can transmit either protumoral or antitumoral signals to glioma cells. Here, we performed bulk RNA sequencing and single-cell RNA sequencing on samples from patients with GBM, which revealed increased ATP synthase expression and oxidative phosphorylation activity in TAMs located in the tumor core relative to the tumor periphery. Both in vitro and in vivo models displayed similar trends of augmented TAM mitochondrial activity, along with elevated mitochondrial fission, glucose uptake, mitochondrial membrane potential, and extracellular ATP (eATP) production by TAMs in the presence of GBM cells. Tumor-secreted factors, including GM-CSF, induced the increase in TAM eATP production. Elevated eATP in the GBM microenvironment promoted glioma growth and invasion by activating the P2X purinoceptor 7 (P2X7R) on glioma cells. Inhibition of the eATP–P2X7R axis attenuated tumor cell viability in vitro and reduced tumor size and prolonged survival in glioma-bearing mouse models. Overall, this study revealed elevated TAM-derived eATP in GBM and provided the basis for targeting the eATP–P2X7R signaling axis as a therapeutic strategy in GBM. Significance: Glioblastoma-mediated metabolic reprogramming in tumor-associated microglia increases ATP secretion that supports cancer cell proliferation and invasion by activating P2X7R, which can be inhibited to attenuate tumor growth.
{"title":"Tumor-Associated Microglia Secrete Extracellular ATP to Support Glioblastoma Progression","authors":"Caren Yu-Ju Wu, Yiyun Chen, Ya-Jui Lin, Kuo-Chen Wei, Kwang-Yu Chang, Li-Ying Feng, Ko-Ting Chen, Gordon Li, Alexander Liang Ren, Ryan Takeo Nitta, Janet Yuling Wu, Kwang Bog Cho, Ayush Pant, John Choi, Crystal L. Mackall, Lily H. Kim, An-Chih Wu, Jian-Ying Chuang, Chiung-Yin Huang, Christopher M. Jackson, Pin-Yuan Chen, Michael Lim","doi":"10.1158/0008-5472.can-24-0018","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-0018","url":null,"abstract":"Glioblastoma (GBM) is a highly aggressive brain tumor with poor prognosis and high recurrence rates. The complex immune microenvironment of GBM is highly infiltrated by tumor-associated microglia and macrophages (TAM). TAMs are known to be heterogeneous in their functional and metabolic states and can transmit either protumoral or antitumoral signals to glioma cells. Here, we performed bulk RNA sequencing and single-cell RNA sequencing on samples from patients with GBM, which revealed increased ATP synthase expression and oxidative phosphorylation activity in TAMs located in the tumor core relative to the tumor periphery. Both in vitro and in vivo models displayed similar trends of augmented TAM mitochondrial activity, along with elevated mitochondrial fission, glucose uptake, mitochondrial membrane potential, and extracellular ATP (eATP) production by TAMs in the presence of GBM cells. Tumor-secreted factors, including GM-CSF, induced the increase in TAM eATP production. Elevated eATP in the GBM microenvironment promoted glioma growth and invasion by activating the P2X purinoceptor 7 (P2X7R) on glioma cells. Inhibition of the eATP–P2X7R axis attenuated tumor cell viability in vitro and reduced tumor size and prolonged survival in glioma-bearing mouse models. Overall, this study revealed elevated TAM-derived eATP in GBM and provided the basis for targeting the eATP–P2X7R signaling axis as a therapeutic strategy in GBM. Significance: Glioblastoma-mediated metabolic reprogramming in tumor-associated microglia increases ATP secretion that supports cancer cell proliferation and invasion by activating P2X7R, which can be inhibited to attenuate tumor growth.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"78 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142760334","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tumor Evolution Reconstruction Is Heavily Influenced by Algorithmic and Experimental Choices. 肿瘤演化重建在很大程度上受到算法和实验选择的影响。
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-12-02 DOI: 10.1158/0008-5472.CAN-24-3530
Rija Zaidi, Simone Zaccaria

Tumor progression is an evolutionary process during which cells acquire distinct genetic alterations. Several cancer evolutionary studies reconstruct this evolutionary process by applying bulk DNA sequencing to a tumor sample to infer the presence of genetic alterations using various tumor evolutionary algorithms. Through a comprehensive benchmarking effort of these algorithms, a recent study by Salcedo and colleagues found that algorithmic and experimental choices are the main drivers of the accuracy of tumor evolution reconstruction, shedding new light on interpreting previous studies and suggesting a useful path forward for the research community.

肿瘤进展是一个进化过程,在这一过程中,细胞会获得不同的基因改变。一些癌症进化研究通过对肿瘤样本进行大量 DNA 测序,利用各种肿瘤进化算法推断基因改变的存在,从而重建这一进化过程。通过对这些算法进行全面的基准测试,Salcedo 及其同事最近的一项研究发现,算法和实验选择是影响肿瘤进化重建准确性的主要因素,这为解读以前的研究提供了新的思路,并为研究界提出了一条有用的前进道路。
{"title":"Tumor Evolution Reconstruction Is Heavily Influenced by Algorithmic and Experimental Choices.","authors":"Rija Zaidi, Simone Zaccaria","doi":"10.1158/0008-5472.CAN-24-3530","DOIUrl":"10.1158/0008-5472.CAN-24-3530","url":null,"abstract":"<p><p>Tumor progression is an evolutionary process during which cells acquire distinct genetic alterations. Several cancer evolutionary studies reconstruct this evolutionary process by applying bulk DNA sequencing to a tumor sample to infer the presence of genetic alterations using various tumor evolutionary algorithms. Through a comprehensive benchmarking effort of these algorithms, a recent study by Salcedo and colleagues found that algorithmic and experimental choices are the main drivers of the accuracy of tumor evolution reconstruction, shedding new light on interpreting previous studies and suggesting a useful path forward for the research community.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"3921-3923"},"PeriodicalIF":12.5,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142364509","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chromatin Helicase CHD6 Establishes Pro-inflammatory Enhancers and is a Synthetic Lethal Target in FH-Deficient Renal Cell Carcinoma. 染色质螺旋酶 CHD6 在 FH 缺失的肾细胞癌中建立促炎增强子并成为合成致命靶点
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-26 DOI: 10.1158/0008-5472.CAN-24-0787
Juan Jin, Jun Luo, Xiaodong Jin, Kiat Shenq Lim, Yang He, Jiawei Ding, Yan Shen, Yuchen Hou, Hanqing Liu, Xiaoyu Zhu, Jing Zhao, Wenjie Zhou, Hai Huang, Yi Gao, Jun Xiao, Hongchao He, Qunyi Li, Lianxin Liu, Li Chen, Qiang He, Chuanjie Zhang

Fumarate hydratase (FH) deficiency causes hereditary leiomyomatosis and renal cell carcinoma (RCC). FH-deficient tumors lack effective therapeutic options. Here, we utilized an epigenetic-focused single-guide RNA library to elucidate potential drug targets in FH-deficient tumors. The screen identified chromodomain helicase DNA binding protein 6 (CHD6) as an essential regulator of the growth of FH-mutated RCC. Mechanically, FH loss induced fumarate-mediated succinylation and inactivation of KEAP1, blocking subsequent ubiquitin-proteasome degradation of CHD6. Stabilized CHD6 formed a complex with p65 to establish pro-inflammatory enhancers and thereby regulate NF-κB-mediated transcription. Moreover, CHD6 recruited mSWI/SNF ATPases to maintain chromatin accessibility at CHD6-bound enhancers. The PROTAC degrader of SMARCA2/4 AU-15330 effectively abolished structures of cis-regulatory elements bound by CHD6 and suppressed the growth of FH-mutated, but not FH-intact, RCC in vivo. Collectively, these data indicate that CHD6 is a molecular bridge between FH deficiency and pro-inflammatory enhancers assembly that endows FH-deficient tumors with epigenetic vulnerabilities.

富马酸氢化酶(FH)缺乏症会导致遗传性乳糜泻和肾细胞癌(RCC)。缺乏富马酸氢化酶的肿瘤缺乏有效的治疗方案。在这里,我们利用以表观遗传学为重点的单导RNA文库来阐明FH缺陷型肿瘤的潜在药物靶点。筛选结果表明,染色体结构域螺旋酶DNA结合蛋白6(CHD6)是FH突变RCC生长的重要调节因子。从机制上讲,FH缺失诱导富马酸介导的琥珀酰化和KEAP1失活,阻止了CHD6随后的泛素蛋白酶体降解。稳定的 CHD6 与 p65 形成复合物,以建立促炎增强子,从而调节 NF-κB 介导的转录。此外,CHD6 还招募 mSWI/SNF ATPases 来维持 CHD6 结合的增强子的染色质可及性。SMARCA2/4的PROTAC降解剂AU-15330有效地消除了CHD6结合的顺式调控元件的结构,并抑制了体内FH突变的RCC的生长,而不是FH未突变的RCC的生长。这些数据共同表明,CHD6是FH缺乏与促炎增强子组装之间的分子桥梁,它赋予了FH缺乏的肿瘤以表观遗传脆弱性。
{"title":"Chromatin Helicase CHD6 Establishes Pro-inflammatory Enhancers and is a Synthetic Lethal Target in FH-Deficient Renal Cell Carcinoma.","authors":"Juan Jin, Jun Luo, Xiaodong Jin, Kiat Shenq Lim, Yang He, Jiawei Ding, Yan Shen, Yuchen Hou, Hanqing Liu, Xiaoyu Zhu, Jing Zhao, Wenjie Zhou, Hai Huang, Yi Gao, Jun Xiao, Hongchao He, Qunyi Li, Lianxin Liu, Li Chen, Qiang He, Chuanjie Zhang","doi":"10.1158/0008-5472.CAN-24-0787","DOIUrl":"https://doi.org/10.1158/0008-5472.CAN-24-0787","url":null,"abstract":"<p><p>Fumarate hydratase (FH) deficiency causes hereditary leiomyomatosis and renal cell carcinoma (RCC). FH-deficient tumors lack effective therapeutic options. Here, we utilized an epigenetic-focused single-guide RNA library to elucidate potential drug targets in FH-deficient tumors. The screen identified chromodomain helicase DNA binding protein 6 (CHD6) as an essential regulator of the growth of FH-mutated RCC. Mechanically, FH loss induced fumarate-mediated succinylation and inactivation of KEAP1, blocking subsequent ubiquitin-proteasome degradation of CHD6. Stabilized CHD6 formed a complex with p65 to establish pro-inflammatory enhancers and thereby regulate NF-κB-mediated transcription. Moreover, CHD6 recruited mSWI/SNF ATPases to maintain chromatin accessibility at CHD6-bound enhancers. The PROTAC degrader of SMARCA2/4 AU-15330 effectively abolished structures of cis-regulatory elements bound by CHD6 and suppressed the growth of FH-mutated, but not FH-intact, RCC in vivo. Collectively, these data indicate that CHD6 is a molecular bridge between FH deficiency and pro-inflammatory enhancers assembly that endows FH-deficient tumors with epigenetic vulnerabilities.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":""},"PeriodicalIF":12.5,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142715406","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Adapt or Perish: Efficient Selenocysteine Insertion is Critical for Metastasizing Cancer Cells. 适应或灭亡:高效的硒半胱氨酸插入对转移癌细胞至关重要
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-26 DOI: 10.1158/0008-5472.CAN-24-4442
Namgyu Lee, Dohoon Kim

During metastasis, cancer cells detach from the primary tumor, circulate through the bloodstream, and establish themselves at distant sites, facing increased levels of reactive oxygen species (ROS) that act as significant barriers to metastatic progression. Adapting to and surviving in these high-ROS environments is thus crucial for successful metastasis. A recent study by Nease and colleagues identified FTSJ1 as the methyltransferase responsible for methylation of the U34 position wobble uridine modification of selenocysteine (Sec) tRNA. This methylation enables efficient Sec insertion, leading to increased translation of a subset of stress-responsive selenoproteins that combat the oxidative stress encountered during the metastatic process. This study establishes FTSJ1 as an essential redox regulator during metastasis through its role in enhancing Sec insertion efficiency, and introduces a potential therapeutic strategy against metastasis.

在转移过程中,癌细胞脱离原发肿瘤,在血液中循环,并在遥远的地方立足,面临着活性氧(ROS)水平升高的问题,而活性氧是转移进展的重要障碍。因此,适应这些高活性氧环境并在其中存活是成功转移的关键。Nease 及其同事最近的一项研究发现,FTSJ1 是负责硒半胱氨酸(Sec)tRNA U34 位摆动尿苷修饰甲基化的甲基转移酶。这种甲基化使 Sec 得以有效插入,从而增加了应激反应硒蛋白子集的翻译,以对抗转移过程中遇到的氧化应激。这项研究通过 FTSJ1 在提高 Sec 插入效率方面的作用,确定了它是转移过程中一个重要的氧化还原调节因子,并提出了一种潜在的转移治疗策略。
{"title":"Adapt or Perish: Efficient Selenocysteine Insertion is Critical for Metastasizing Cancer Cells.","authors":"Namgyu Lee, Dohoon Kim","doi":"10.1158/0008-5472.CAN-24-4442","DOIUrl":"https://doi.org/10.1158/0008-5472.CAN-24-4442","url":null,"abstract":"<p><p>During metastasis, cancer cells detach from the primary tumor, circulate through the bloodstream, and establish themselves at distant sites, facing increased levels of reactive oxygen species (ROS) that act as significant barriers to metastatic progression. Adapting to and surviving in these high-ROS environments is thus crucial for successful metastasis. A recent study by Nease and colleagues identified FTSJ1 as the methyltransferase responsible for methylation of the U34 position wobble uridine modification of selenocysteine (Sec) tRNA. This methylation enables efficient Sec insertion, leading to increased translation of a subset of stress-responsive selenoproteins that combat the oxidative stress encountered during the metastatic process. This study establishes FTSJ1 as an essential redox regulator during metastasis through its role in enhancing Sec insertion efficiency, and introduces a potential therapeutic strategy against metastasis.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":""},"PeriodicalIF":12.5,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142715268","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Breast Cancer Subtype-Specific Organotropism is Dictated by FOXF2-Regulated Metastatic Dormancy and Recovery. 乳腺癌亚型特异性器官转移受 FOXF2 调节的转移蛰伏和恢复的支配
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-26 DOI: 10.1158/0008-5472.CAN-24-0479
Wen-Jing Jiang, Tian-Hao Zhou, Huan-Jing Huang, Lin-Sen Li, Hao Tan, Rui Zhang, Qing-Shan Wang, Yu-Mei Feng

Breast cancer subtypes display different metastatic organotropism. Identification of the mechanisms underlying subtype-specific organotropism could help uncover potential approaches to prevent and treat metastasis. Herein, we found that FOXF2 promoted the seeding and proliferative recovery from dormancy of luminal breast cancer (LumBC) and basal-like breast cancer (BLBC) cells in the bone by activating the NF-κB and BMP signaling pathways. Conversely, FOXF2 suppressed the seeding and proliferative recovery of BLBC cells in the lung by repressing the TGF-β signaling pathway. FOXF2 directly upregulated RelA/p65 transcription and expression in LumBC and BLBC cells by binding to the RELA proximal promoter region, and RelA/p65 bound to the FOXF2 proximal promoter region to upregulate expression, forming a positive feedback loop. Targeting the NF-κB pathway efficiently prevented the metastasis of FOXF2-overexpressing breast cancer cells to the bone, while inhibiting TGF-β signaling blocked the metastasis of BLBC with low FOXF2 expression to the lung. These findings uncover critical mechanisms of breast cancer subtype-specific organotropism and provide insight into precision assessment and treatment strategies.

乳腺癌亚型表现出不同的转移性器官移动性。鉴定亚型特异性器官向性的机制有助于发现预防和治疗转移的潜在方法。在本文中,我们发现FOXF2通过激活NF-κB和BMP信号通路,促进腔隙性乳腺癌(LumBC)和基底样乳腺癌(BLBC)细胞在骨中播种并从休眠中恢复增殖。相反,FOXF2通过抑制TGF-β信号通路,抑制了BLBC细胞在肺部的播种和增殖恢复。FOXF2通过与RELA近端启动子区域结合,直接上调LumBC和BLBC细胞中RelA/p65的转录和表达,而RelA/p65则与FOXF2近端启动子区域结合,上调表达,形成正反馈回路。靶向NF-κB通路可有效阻止FOXF2表达量低的乳腺癌细胞向骨转移,而抑制TGF-β信号传导则可阻止FOXF2表达量低的BLBC向肺部转移。这些发现揭示了乳腺癌亚型特异性器官转移的关键机制,为精准评估和治疗策略提供了启示。
{"title":"Breast Cancer Subtype-Specific Organotropism is Dictated by FOXF2-Regulated Metastatic Dormancy and Recovery.","authors":"Wen-Jing Jiang, Tian-Hao Zhou, Huan-Jing Huang, Lin-Sen Li, Hao Tan, Rui Zhang, Qing-Shan Wang, Yu-Mei Feng","doi":"10.1158/0008-5472.CAN-24-0479","DOIUrl":"https://doi.org/10.1158/0008-5472.CAN-24-0479","url":null,"abstract":"<p><p>Breast cancer subtypes display different metastatic organotropism. Identification of the mechanisms underlying subtype-specific organotropism could help uncover potential approaches to prevent and treat metastasis. Herein, we found that FOXF2 promoted the seeding and proliferative recovery from dormancy of luminal breast cancer (LumBC) and basal-like breast cancer (BLBC) cells in the bone by activating the NF-κB and BMP signaling pathways. Conversely, FOXF2 suppressed the seeding and proliferative recovery of BLBC cells in the lung by repressing the TGF-β signaling pathway. FOXF2 directly upregulated RelA/p65 transcription and expression in LumBC and BLBC cells by binding to the RELA proximal promoter region, and RelA/p65 bound to the FOXF2 proximal promoter region to upregulate expression, forming a positive feedback loop. Targeting the NF-κB pathway efficiently prevented the metastasis of FOXF2-overexpressing breast cancer cells to the bone, while inhibiting TGF-β signaling blocked the metastasis of BLBC with low FOXF2 expression to the lung. These findings uncover critical mechanisms of breast cancer subtype-specific organotropism and provide insight into precision assessment and treatment strategies.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":""},"PeriodicalIF":12.5,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142715271","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fungal Influences on Cancer Initiation, Progression, and Response to Treatment. 真菌对癌症发生、发展和治疗反应的影响
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-26 DOI: 10.1158/0008-5472.CAN-24-1609
Hazrat Bilal, Muhammad Nadeem Khan, Sabir Khan, Muhammad Shafiq, Wenjie Fang, Yuebin Zeng, Yangzhong Guo, Xiaohui Li, Bing Zhao, Qiao-Li Lv, Bin Xu

Fungal dysbiosis is increasingly recognized as a key factor in cancer, influencing tumor initiation, progression, and treatment outcomes. This review explores the role of fungi in carcinogenesis, with a focus on mechanisms such as immunomodulation, inflammation induction, tumor microenvironment remodeling, and interkingdom interactions. Fungal metabolites are involved in oncogenesis, and antifungals can interact with anticancer drug, including eliciting potential adverse effects and influencing immune responses. Furthermore, mycobiota profiles have potential as diagnostic and prognostic biomarkers, emphasizing their clinical relevance. The interplay between fungi and cancer therapies can impact drug resistance, therapeutic efficacy, and risk of invasive fungal infections associated with targeted therapies. Finally, emerging strategies for modulating mycobiota in cancer care are promising approaches to improve patient outcomes.

真菌菌群失调越来越被认为是癌症的一个关键因素,它影响着肿瘤的发生、发展和治疗效果。这篇综述探讨了真菌在致癌过程中的作用,重点关注免疫调节、炎症诱导、肿瘤微环境重塑和王国间相互作用等机制。真菌代谢产物参与肿瘤发生,抗真菌药物可与抗癌药物相互作用,包括引起潜在的不良反应和影响免疫反应。此外,真菌生物群图谱有可能成为诊断和预后的生物标志物,强调了其临床相关性。真菌与癌症疗法之间的相互作用会影响耐药性、疗效以及与靶向疗法相关的侵袭性真菌感染风险。最后,在癌症治疗中调节真菌生物群的新策略是改善患者预后的有希望的方法。
{"title":"Fungal Influences on Cancer Initiation, Progression, and Response to Treatment.","authors":"Hazrat Bilal, Muhammad Nadeem Khan, Sabir Khan, Muhammad Shafiq, Wenjie Fang, Yuebin Zeng, Yangzhong Guo, Xiaohui Li, Bing Zhao, Qiao-Li Lv, Bin Xu","doi":"10.1158/0008-5472.CAN-24-1609","DOIUrl":"https://doi.org/10.1158/0008-5472.CAN-24-1609","url":null,"abstract":"<p><p>Fungal dysbiosis is increasingly recognized as a key factor in cancer, influencing tumor initiation, progression, and treatment outcomes. This review explores the role of fungi in carcinogenesis, with a focus on mechanisms such as immunomodulation, inflammation induction, tumor microenvironment remodeling, and interkingdom interactions. Fungal metabolites are involved in oncogenesis, and antifungals can interact with anticancer drug, including eliciting potential adverse effects and influencing immune responses. Furthermore, mycobiota profiles have potential as diagnostic and prognostic biomarkers, emphasizing their clinical relevance. The interplay between fungi and cancer therapies can impact drug resistance, therapeutic efficacy, and risk of invasive fungal infections associated with targeted therapies. Finally, emerging strategies for modulating mycobiota in cancer care are promising approaches to improve patient outcomes.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":""},"PeriodicalIF":12.5,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142715426","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Potent, Selective, Small-Molecule Inhibitor of DHX9 Abrogates Proliferation of Microsatellite Instable Cancers with Deficient Mismatch Repair. 一种强效、选择性小分子 DHX9 抑制剂可抑制错配修复缺陷的微卫星不稳定性癌症的增殖
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-26 DOI: 10.1158/0008-5472.CAN-24-0397
Jennifer Castro, Matthew H Daniels, David Brennan, Brian Johnston, Deepali Gotur, Young-Tae Lee, Kevin E Knockenhauer, Chuang Lu, Jie Wu, Sunaina Nayak, Cindy Collins, Rishabh Bansal, Shane M Buker, April Case, Julie Liu, Shihua Yao, Brian A Sparling, E Allen Sickmier, Serena J Silver, Stephen J Blakemore, P Ann Boriack-Sjodin, Kenneth W Duncan, Scott Ribich, Robert A Copeland

DHX9 is a multifunctional DExH-box RNA helicase with important roles in the regulation of transcription, translation, and maintenance of genome stability. Elevated expression of DHX9 is evident in multiple cancer types, including colorectal cancer (CRC). Microsatellite instable-high (MSI-H) tumors with deficient mismatch repair (dMMR) display a strong dependence on DHX9, making this helicase an attractive target for oncology drug discovery. In this report, we show that DHX9 knockdown increased RNA/DNA secondary structures and replication stress, resulting in cell cycle arrest and the onset of apoptosis in cancer cells with MSI-H/dMMR. ATX968 was identified as a potent and selective inhibitor of DHX9 helicase activity. Chemical inhibition of DHX9 enzymatic activity elicited similar selective effects on cell proliferation as seen with genetic knockdown. In addition, ATX968 induced robust and durable responses in an MSI-H/dMMR xenograft model but not in a microsatellite stable (MSS)/proficient mismatch repair (pMMR) model. These preclinical data validate DHX9 as a target for the treatment of patients with MSI-H/dMMR. Additionally, this potent and selective inhibitor of DHX9 provides a valuable tool with which to further explore the effects of inhibition of DHX9 enzymatic activity on the proliferation of cancer cells in vitro and in vivo.

DHX9 是一种多功能 DExH-box RNA 螺旋酶,在调控转录、翻译和维持基因组稳定性方面发挥着重要作用。在包括结直肠癌(CRC)在内的多种癌症类型中,DHX9 的表达明显升高。具有错配修复缺陷(dMMR)的微卫星不稳定性高(MSI-H)肿瘤对 DHX9 有很强的依赖性,这使得这种螺旋酶成为肿瘤药物发现的一个有吸引力的靶点。在本报告中,我们发现 DHX9 基因敲除会增加 RNA/DNA 二级结构和复制应激,导致 MSI-H/dMMR 癌细胞的细胞周期停滞和凋亡。ATX968 被鉴定为 DHX9 螺旋酶活性的强效选择性抑制剂。化学抑制 DHX9 酶活性对细胞增殖的选择性影响与基因敲除类似。此外,ATX968还能在MSI-H/dMMR异种移植模型中诱导稳健持久的反应,但在微卫星稳定(MSS)/错配修复能力强(pMMR)模型中却不能。这些临床前数据验证了 DHX9 是治疗 MSI-H/dMMR 患者的靶点。此外,这种强效的 DHX9 选择性抑制剂为进一步探索抑制 DHX9 酶活性对体外和体内癌细胞增殖的影响提供了宝贵的工具。
{"title":"A Potent, Selective, Small-Molecule Inhibitor of DHX9 Abrogates Proliferation of Microsatellite Instable Cancers with Deficient Mismatch Repair.","authors":"Jennifer Castro, Matthew H Daniels, David Brennan, Brian Johnston, Deepali Gotur, Young-Tae Lee, Kevin E Knockenhauer, Chuang Lu, Jie Wu, Sunaina Nayak, Cindy Collins, Rishabh Bansal, Shane M Buker, April Case, Julie Liu, Shihua Yao, Brian A Sparling, E Allen Sickmier, Serena J Silver, Stephen J Blakemore, P Ann Boriack-Sjodin, Kenneth W Duncan, Scott Ribich, Robert A Copeland","doi":"10.1158/0008-5472.CAN-24-0397","DOIUrl":"https://doi.org/10.1158/0008-5472.CAN-24-0397","url":null,"abstract":"<p><p>DHX9 is a multifunctional DExH-box RNA helicase with important roles in the regulation of transcription, translation, and maintenance of genome stability. Elevated expression of DHX9 is evident in multiple cancer types, including colorectal cancer (CRC). Microsatellite instable-high (MSI-H) tumors with deficient mismatch repair (dMMR) display a strong dependence on DHX9, making this helicase an attractive target for oncology drug discovery. In this report, we show that DHX9 knockdown increased RNA/DNA secondary structures and replication stress, resulting in cell cycle arrest and the onset of apoptosis in cancer cells with MSI-H/dMMR. ATX968 was identified as a potent and selective inhibitor of DHX9 helicase activity. Chemical inhibition of DHX9 enzymatic activity elicited similar selective effects on cell proliferation as seen with genetic knockdown. In addition, ATX968 induced robust and durable responses in an MSI-H/dMMR xenograft model but not in a microsatellite stable (MSS)/proficient mismatch repair (pMMR) model. These preclinical data validate DHX9 as a target for the treatment of patients with MSI-H/dMMR. Additionally, this potent and selective inhibitor of DHX9 provides a valuable tool with which to further explore the effects of inhibition of DHX9 enzymatic activity on the proliferation of cancer cells in vitro and in vivo.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":""},"PeriodicalIF":12.5,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142715324","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mapping of the T Cell Landscape of Biliary Tract Cancer Unravels Anatomical Subtype-Specific Heterogeneity 胆道癌 T 细胞图谱揭示了解剖亚型特异性异质性
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-21 DOI: 10.1158/0008-5472.can-24-1173
Jianhua Nie, Shuyuan Zhang, Ying Guo, Caiqi Liu, Jiaqi Shi, Haotian Wu, Ruisi Na, Yingjian Liang, Shan Yu, Fei Quan, Kun Liu, Mingwei Li, Meng Zhou, Ying Zhao, Xuehan Li, Shengnan Luo, Qian Zhang, Guangyu Wang, Yanqiao Zhang, Yuanfei Yao, Yun Xiao, Sheng Tai, Tongsen Zheng
Biliary tract cancer (BTC), encompassing diseases such as intrahepatic (ICC), extrahepatic cholangiocarcinoma (ECC), and gallbladder cancer (GBC), is not only on the rise but also poses a significant and urgent health threat due to its high malignancy. Genomic differences point to the possibility that these subtypes represent distinct diseases. Elucidation of the specific distribution of T cell subsets, critical to cancer immunity, across these diseases could provide better insights into the unique biology of BTC subtypes and help identify potential precision medicine strategies. To address this, we conducted scRNA-seq and scTCR-seq on CD3+ T cells from 36 samples from 16 BTC patients across all subtypes and analyzed 355 pathological slides to examine the spatial distribution of T cells and tertiary lymphoid structures (TLS). Compared to ICC and GBC, ECC possessed a unique immune profile characterized by T cell exhaustion, elevated CXCL13 expression in CD4+ T helper-like and CD8+CXCL13+ exhausted T cells, more mature TLS, and fewer desert immunophenotypes. Conversely, ICC displayed an inflamed immunophenotype with an enrichment of interferon related pathways and high expression of LGALS1 in activated regulatory T cells, associated with immunosuppression. Inhibition of LGALS1 reduced tumor growth and Treg prevalence in ICC mouse models. Overall, this study unveils T cell diversity across BTC subtypes at the single-cell and spatial level that could open paths for tailored immunotherapies.
胆道癌(BTC)包括肝内胆管癌(ICC)、肝外胆管癌(ECC)和胆囊癌(GBC)等疾病,不仅发病率呈上升趋势,而且因其恶性程度高而对健康构成重大而紧迫的威胁。基因组差异表明,这些亚型可能代表不同的疾病。阐明对癌症免疫至关重要的 T 细胞亚群在这些疾病中的具体分布,可以更好地了解 BTC 亚型的独特生物学特性,并有助于确定潜在的精准医疗策略。为此,我们对来自16名BTC患者的36份样本中的CD3+ T细胞进行了scRNA-seq和scTCR-seq分析,这些样本来自所有亚型,我们还分析了355张病理切片,以研究T细胞和三级淋巴结构(TLS)的空间分布。与 ICC 和 GBC 相比,ECC 具有独特的免疫特征,其特点是 T 细胞衰竭、CD4+ T 辅助细胞样和 CD8+CXCL13+ 衰竭 T 细胞中 CXCL13 表达升高、TLS 更成熟、沙漠免疫表型更少。相反,ICC 显示出炎症免疫表型,干扰素相关通路丰富,活化的调节性 T 细胞中 LGALS1 高表达,这与免疫抑制有关。抑制 LGALS1 可减少 ICC 小鼠模型中的肿瘤生长和 Treg 的流行。总之,这项研究在单细胞和空间水平上揭示了BTC亚型中T细胞的多样性,为定制免疫疗法开辟了道路。
{"title":"Mapping of the T Cell Landscape of Biliary Tract Cancer Unravels Anatomical Subtype-Specific Heterogeneity","authors":"Jianhua Nie, Shuyuan Zhang, Ying Guo, Caiqi Liu, Jiaqi Shi, Haotian Wu, Ruisi Na, Yingjian Liang, Shan Yu, Fei Quan, Kun Liu, Mingwei Li, Meng Zhou, Ying Zhao, Xuehan Li, Shengnan Luo, Qian Zhang, Guangyu Wang, Yanqiao Zhang, Yuanfei Yao, Yun Xiao, Sheng Tai, Tongsen Zheng","doi":"10.1158/0008-5472.can-24-1173","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-1173","url":null,"abstract":"Biliary tract cancer (BTC), encompassing diseases such as intrahepatic (ICC), extrahepatic cholangiocarcinoma (ECC), and gallbladder cancer (GBC), is not only on the rise but also poses a significant and urgent health threat due to its high malignancy. Genomic differences point to the possibility that these subtypes represent distinct diseases. Elucidation of the specific distribution of T cell subsets, critical to cancer immunity, across these diseases could provide better insights into the unique biology of BTC subtypes and help identify potential precision medicine strategies. To address this, we conducted scRNA-seq and scTCR-seq on CD3+ T cells from 36 samples from 16 BTC patients across all subtypes and analyzed 355 pathological slides to examine the spatial distribution of T cells and tertiary lymphoid structures (TLS). Compared to ICC and GBC, ECC possessed a unique immune profile characterized by T cell exhaustion, elevated CXCL13 expression in CD4+ T helper-like and CD8+CXCL13+ exhausted T cells, more mature TLS, and fewer desert immunophenotypes. Conversely, ICC displayed an inflamed immunophenotype with an enrichment of interferon related pathways and high expression of LGALS1 in activated regulatory T cells, associated with immunosuppression. Inhibition of LGALS1 reduced tumor growth and Treg prevalence in ICC mouse models. Overall, this study unveils T cell diversity across BTC subtypes at the single-cell and spatial level that could open paths for tailored immunotherapies.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"71 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142684314","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer research
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