首页 > 最新文献

Molecular Cancer Research最新文献

英文 中文
PCSK5M452I is a recessive hypomorph exclusive to MCF10DCIS.com cells. PCSK5M452I是MCF10DCIS.com细胞独有的隐性低形态。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-28 DOI: 10.1158/1541-7786.MCR-25-0211
Taylor Marohl, Kristen A Atkins, Lixin Wang, Kevin A Janes

The most widely used cell line for studying ductal carcinoma in situ (DCIS) premalignancy is the transformed breast epithelial cell line, MCF10DCIS.com. During its original clonal isolation and selection, MCF10DCIS.com acquired a heterozygous M452I mutation in the proprotein convertase PCSK5, which has never been reported in any human cancer. The mutation is noteworthy because PCSK5 matures GDF11, a TGFβ-superfamily ligand that suppresses progression of triple-negative breast cancer. We asked here whether PCSK5M452I and its activity toward GDF11 might contribute to the unique properties of MCF10DCIS.com. Using an optimized in-cell GDF11 maturation assay, we found that overexpressed PCSK5M452I was measurably active but at a fraction of the wildtype enzyme. In a PCSK5-/- clone of MCF10DCIS.com reconstituted with different PCSK5 alleles, PCSK5M452I was mildly defective in anterograde transport. However, the multicellular organization of PCSK5M452I addback cells in 3D matrigel cultures was significantly less circumscribed than wildtype and indistinguishable from a PCSK5T288P null allele. Growth of intraductal MCF10DCIS.com xenografts was similarly impaired along with the frequency of comedo necrosis and stromal activation. In no setting did PCSK5M452I exhibit gain-of-function activity, leading us to conclude that it is hypomorphic and thus compensated by the remaining wildtype allele in MCF10DCIS.com. Implications: This work reassures that an exotic PCSK5 mutation is not responsible for the salient characteristics of the MCF10DCIS.com cell line.

用于研究导管原位癌(DCIS)恶性前病变最广泛的细胞系是转化乳腺上皮细胞系MCF10DCIS.com。在最初的克隆分离和选择过程中,MCF10DCIS.com获得了蛋白转化酶PCSK5的杂合M452I突变,该突变从未在任何人类癌症中报道过。该突变值得注意,因为PCSK5使GDF11成熟,GDF11是一种tgf β超家族配体,可抑制三阴性乳腺癌的进展。我们在这里询问PCSK5M452I及其对GDF11的活性是否有助于MCF10DCIS.com的独特性质。通过优化的细胞内GDF11成熟实验,我们发现过表达的PCSK5M452I具有可测量的活性,但只具有一小部分野生型酶。在不同PCSK5等位基因重组的MCF10DCIS.com的PCSK5-/-克隆中,PCSK5M452I在顺行运输中存在轻度缺陷。然而,在3D基质培养中,PCSK5M452I addback细胞的多细胞组织明显比野生型少,并且与PCSK5T288P零等位基因无法区分。导管内MCF10DCIS.com异种移植物的生长也受到类似的损害,同时出现粉刺坏死和基质激活的频率也有所降低。在任何情况下,PCSK5M452I都没有表现出功能获得活性,这使我们得出结论,它是半胚性的,因此由MCF10DCIS.com中剩余的野生型等位基因补偿。结论:这项研究再次证实了一种外来的PCSK5突变并不是MCF10DCIS.com细胞系显著特征的原因。
{"title":"PCSK5M452I is a recessive hypomorph exclusive to MCF10DCIS.com cells.","authors":"Taylor Marohl, Kristen A Atkins, Lixin Wang, Kevin A Janes","doi":"10.1158/1541-7786.MCR-25-0211","DOIUrl":"10.1158/1541-7786.MCR-25-0211","url":null,"abstract":"<p><p>The most widely used cell line for studying ductal carcinoma in situ (DCIS) premalignancy is the transformed breast epithelial cell line, MCF10DCIS.com. During its original clonal isolation and selection, MCF10DCIS.com acquired a heterozygous M452I mutation in the proprotein convertase PCSK5, which has never been reported in any human cancer. The mutation is noteworthy because PCSK5 matures GDF11, a TGFβ-superfamily ligand that suppresses progression of triple-negative breast cancer. We asked here whether PCSK5M452I and its activity toward GDF11 might contribute to the unique properties of MCF10DCIS.com. Using an optimized in-cell GDF11 maturation assay, we found that overexpressed PCSK5M452I was measurably active but at a fraction of the wildtype enzyme. In a PCSK5-/- clone of MCF10DCIS.com reconstituted with different PCSK5 alleles, PCSK5M452I was mildly defective in anterograde transport. However, the multicellular organization of PCSK5M452I addback cells in 3D matrigel cultures was significantly less circumscribed than wildtype and indistinguishable from a PCSK5T288P null allele. Growth of intraductal MCF10DCIS.com xenografts was similarly impaired along with the frequency of comedo necrosis and stromal activation. In no setting did PCSK5M452I exhibit gain-of-function activity, leading us to conclude that it is hypomorphic and thus compensated by the remaining wildtype allele in MCF10DCIS.com. Implications: This work reassures that an exotic PCSK5 mutation is not responsible for the salient characteristics of the MCF10DCIS.com cell line.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12671455/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145377925","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
Bone Metastatic Progression of Prostate Cancer is Regulated by TRIM28-LDHA Mediated Metabolism. TRIM28-LDHA介导的代谢调节前列腺癌骨转移进展
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-17 DOI: 10.1158/1541-7786.MCR-25-0676
Miyeong Kim, Han Cong, Ryan Goettl, Jinpeng Liu, Ka-Wing Fong

Castration-resistant prostate cancer (CRPC), an advanced stage of prostate cancer (PCa), often leads to fatal bone metastasis. The vast majority of PCa patients who present with bone metastases suffer from bone lesions and other complications. Androgen receptor (AR) inhibitors, while improved, lack curative efficacy, necessitating an urgent demand for the development of innovative therapeutic strategies. TRIM28, also known as KAP1, is a transcription factor regulated by site-specific phosphorylation. Our recent study demonstrated that RSK1 is the protein kinase that directly phosphorylates TRIM28 at S473; as such, pS473-TRIM28 promotes the transcriptional activation of its gene targets. In this study, we reveal that TRIM28 S473 phosphorylation is readily detected in CRPC bone metastases, which is consistent with the previous report that RSK kinase is activated in PCa bone metastases. Using bioinformatic and genomic analysis, we uncovered that lactate dehydrogenase A (LDHA) is a novel TRIM28-induced gene in bone metastatic PCa. TRIM28 promotes the transcriptional activation of LDHA in a pS473-TRIM28 dependent manner. As such, TRIM28 is involved in LDH-related activities including lactate production and glycolysis. We also demonstrate that the TRIM28-LDHA axis is required for prostate tumor progression using an orthotopic bone injection model. Lastly, the application of an LDH inhibitor mitigates PCa development in bone. In summary, our study reveals an important role of the TRIM28-LDHA axis in PCa progression in bone, which may be targeted to mitigate the disease in the metastasis stage. Implications: TRIM28 upregulates LDHA and glycolysis, propelling prostate tumors in bone; pharmacologic LDH blockade mitigates disease.

去势抵抗性前列腺癌(CRPC)是一种晚期前列腺癌(PCa),常导致致命的骨转移。绝大多数出现骨转移的前列腺癌患者伴有骨病变和其他并发症。雄激素受体(AR)抑制剂虽然有所改善,但缺乏疗效,迫切需要开发创新的治疗策略。TRIM28,又称KAP1,是一种受位点特异性磷酸化调控的转录因子。我们最近的研究表明,RSK1是直接磷酸化TRIM28在S473位点的蛋白激酶;因此,pS473-TRIM28可促进其靶基因的转录激活。在本研究中,我们发现TRIM28 S473磷酸化在CRPC骨转移中很容易检测到,这与之前报道的RSK激酶在PCa骨转移中被激活一致。通过生物信息学和基因组学分析,我们发现乳酸脱氢酶A (LDHA)是骨转移性前列腺癌中一种新的trim28诱导基因。TRIM28以pS473-TRIM28依赖的方式促进LDHA的转录激活。因此,TRIM28参与ldl相关活动,包括乳酸生成和糖酵解。我们还通过原位骨注射模型证明TRIM28-LDHA轴是前列腺肿瘤进展所必需的。最后,LDH抑制剂的应用减轻了骨内PCa的发展。综上所述,我们的研究揭示了TRIM28-LDHA轴在骨内PCa进展中的重要作用,可能在转移期靶向缓解疾病。提示:TRIM28上调LDHA和糖酵解,促进骨内前列腺肿瘤;药理学上LDH阻断可减轻疾病。
{"title":"Bone Metastatic Progression of Prostate Cancer is Regulated by TRIM28-LDHA Mediated Metabolism.","authors":"Miyeong Kim, Han Cong, Ryan Goettl, Jinpeng Liu, Ka-Wing Fong","doi":"10.1158/1541-7786.MCR-25-0676","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-25-0676","url":null,"abstract":"<p><p>Castration-resistant prostate cancer (CRPC), an advanced stage of prostate cancer (PCa), often leads to fatal bone metastasis. The vast majority of PCa patients who present with bone metastases suffer from bone lesions and other complications. Androgen receptor (AR) inhibitors, while improved, lack curative efficacy, necessitating an urgent demand for the development of innovative therapeutic strategies. TRIM28, also known as KAP1, is a transcription factor regulated by site-specific phosphorylation. Our recent study demonstrated that RSK1 is the protein kinase that directly phosphorylates TRIM28 at S473; as such, pS473-TRIM28 promotes the transcriptional activation of its gene targets. In this study, we reveal that TRIM28 S473 phosphorylation is readily detected in CRPC bone metastases, which is consistent with the previous report that RSK kinase is activated in PCa bone metastases. Using bioinformatic and genomic analysis, we uncovered that lactate dehydrogenase A (LDHA) is a novel TRIM28-induced gene in bone metastatic PCa. TRIM28 promotes the transcriptional activation of LDHA in a pS473-TRIM28 dependent manner. As such, TRIM28 is involved in LDH-related activities including lactate production and glycolysis. We also demonstrate that the TRIM28-LDHA axis is required for prostate tumor progression using an orthotopic bone injection model. Lastly, the application of an LDH inhibitor mitigates PCa development in bone. In summary, our study reveals an important role of the TRIM28-LDHA axis in PCa progression in bone, which may be targeted to mitigate the disease in the metastasis stage. Implications: TRIM28 upregulates LDHA and glycolysis, propelling prostate tumors in bone; pharmacologic LDH blockade mitigates disease.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145308513","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
RAS mutation-specific responses to paralog- and state-selective RAS inhibitors. 对平行和状态选择性RAS抑制剂的RAS突变特异性反应。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-15 DOI: 10.1158/1541-7786.MCR-25-0319
Beau Baars, Ana Orive-Ramos, Matthew J Emmett, Bijaya Gaire, Mathieu Desaunay, Ziyue Kou, Guangyan Li, Christos Adamopoulos, Stuart A Aaronson, Shaomeng Wang, William R Sellers, Tiphaine Martin, Evripidis Gavathiotis, Poulikos I Poulikakos

A high therapeutic index, defined as potent inhibition of oncogenic signaling in tumor cells with minimal effects on normal cells, is critical for effective cancer therapies. Recent advances have introduced diverse RAS-targeting inhibitors, including mutant-specific inhibitors such as KRAS G12C and KRAS G12D, as well as paralog- and state-selective inhibitors. Non-mutant-specific RAS inhibition can be achieved by: (1) guanine nucleotide exchange-OFF inhibitors that indirectly inactivate RAS by targeting SHP2 or SOS1, (2) KRAS-OFF inhibitors that spare NRAS and HRAS, and (3) active-state RAS(ON) inhibitors that directly block binding of effector RAF. However, the signaling inhibition index (SII)-the differential suppression of oncogenic signaling between RAS-mutant and normal cells-remains poorly defined for these approaches. We evaluated the SII for state- and paralog-selective RAS inhibitors across diverse RAS-mutant and RAS-wild-type models. Guanine nucleotide exchange-OFF inhibitors exhibited neutral or negative values, with reduced MAPK suppression in KRAS G12X cells compared to wild-type cells. KRAS G13D models, especially with NF1 loss, showed low sensitivity. SHP2 plus MEK inhibition resulted in low selectivity, and RAS Q61X models were resistant due to MEK inhibitor-induced NRAS reactivation and altered SHP2 conformations. KRAS-OFF inhibitors demonstrated higher selectivity, while active-state RAS(ON) inhibitors showed broader activity but narrow selectivity. Sensitivity to mutant-specific inhibitors largely overlapped with sensitivity to state-selective agents, suggesting that most RAS-mutant tumors will respond poorly to any currently available RAS inhibitor. Implications: Determining the signaling inhibition index (SII) can inform the design and clinical application of RAS-targeted therapies to improve tumor selectivity and therapeutic outcomes.

高治疗指数,定义为肿瘤细胞中致癌信号的有效抑制,对正常细胞的影响最小,对有效的癌症治疗至关重要。最近的进展已经引入了多种ras靶向抑制剂,包括突变特异性抑制剂,如KRAS G12C和KRAS G12D,以及平行和状态选择性抑制剂。非突变特异性RAS抑制可以通过以下方法实现:(1)鸟嘌呤核苷酸交换- off抑制剂通过靶向SHP2或SOS1间接灭活RAS, (2) KRAS-OFF抑制剂使NRAS和HRAS不受影响,以及(3)活性态RAS(ON)抑制剂直接阻断效应RAF的结合。然而,信号抑制指数(SII)——ras突变细胞和正常细胞对致癌信号的差异抑制——在这些方法中仍然定义不清。我们在不同的RAS突变型和RAS野生型模型中评估了状态选择性和副选择性RAS抑制剂的SII。鸟嘌呤核苷酸交换- off抑制剂表现为中性或负值,与野生型细胞相比,KRAS G12X细胞中的MAPK抑制降低。KRAS G13D模型,尤其是NF1损失模型,灵敏度较低。SHP2 + MEK抑制导致低选择性,RAS Q61X模型由于MEK抑制剂诱导的NRAS再激活和SHP2构象的改变而产生耐药性。KRAS-OFF抑制剂具有较高的选择性,而活性态RAS(ON)抑制剂具有较宽的活性,但选择性较窄。对突变特异性抑制剂的敏感性与对状态选择性药物的敏感性在很大程度上重叠,这表明大多数RAS突变肿瘤对任何目前可用的RAS抑制剂的反应都很差。意义:确定信号抑制指数(SII)可以为ras靶向治疗的设计和临床应用提供信息,以提高肿瘤的选择性和治疗效果。
{"title":"RAS mutation-specific responses to paralog- and state-selective RAS inhibitors.","authors":"Beau Baars, Ana Orive-Ramos, Matthew J Emmett, Bijaya Gaire, Mathieu Desaunay, Ziyue Kou, Guangyan Li, Christos Adamopoulos, Stuart A Aaronson, Shaomeng Wang, William R Sellers, Tiphaine Martin, Evripidis Gavathiotis, Poulikos I Poulikakos","doi":"10.1158/1541-7786.MCR-25-0319","DOIUrl":"10.1158/1541-7786.MCR-25-0319","url":null,"abstract":"<p><p>A high therapeutic index, defined as potent inhibition of oncogenic signaling in tumor cells with minimal effects on normal cells, is critical for effective cancer therapies. Recent advances have introduced diverse RAS-targeting inhibitors, including mutant-specific inhibitors such as KRAS G12C and KRAS G12D, as well as paralog- and state-selective inhibitors. Non-mutant-specific RAS inhibition can be achieved by: (1) guanine nucleotide exchange-OFF inhibitors that indirectly inactivate RAS by targeting SHP2 or SOS1, (2) KRAS-OFF inhibitors that spare NRAS and HRAS, and (3) active-state RAS(ON) inhibitors that directly block binding of effector RAF. However, the signaling inhibition index (SII)-the differential suppression of oncogenic signaling between RAS-mutant and normal cells-remains poorly defined for these approaches. We evaluated the SII for state- and paralog-selective RAS inhibitors across diverse RAS-mutant and RAS-wild-type models. Guanine nucleotide exchange-OFF inhibitors exhibited neutral or negative values, with reduced MAPK suppression in KRAS G12X cells compared to wild-type cells. KRAS G13D models, especially with NF1 loss, showed low sensitivity. SHP2 plus MEK inhibition resulted in low selectivity, and RAS Q61X models were resistant due to MEK inhibitor-induced NRAS reactivation and altered SHP2 conformations. KRAS-OFF inhibitors demonstrated higher selectivity, while active-state RAS(ON) inhibitors showed broader activity but narrow selectivity. Sensitivity to mutant-specific inhibitors largely overlapped with sensitivity to state-selective agents, suggesting that most RAS-mutant tumors will respond poorly to any currently available RAS inhibitor. Implications: Determining the signaling inhibition index (SII) can inform the design and clinical application of RAS-targeted therapies to improve tumor selectivity and therapeutic outcomes.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12710464/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145302532","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
Loss of Regulator of G protein signaling 11 promotes pro-tumorigenic features in pancreatic cancer. G蛋白信号11调节因子的缺失促进胰腺癌的致瘤性特征。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-14 DOI: 10.1158/1541-7786.MCR-25-0144
Tejinder Kaur, Debasis Nayak, Arnav Joshi, Junan Li, Amy Hite, Rajgopal Govindarajan

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy, associated with early metastasis, drug resistance and poor outcomes. We previously demonstrated a putative tumor suppressive role for concentrative nucleoside transporter 1 (CNT1) in PDAC. Here we demonstrate the regulator of G protein signaling (RGS) 11 as a key target of CNT1, with potent tumor suppressive properties in PDAC. Compared to normal human pancreas, RGS11 expression is diminished in human PDAC tissues which correspond with the reduced patient survival times. In addition, quasi-mesenchymal pancreatic tumor cell lines with accelerated growth, metastatic propensity, and innate resistance to nucleoside analogs showed relatively lower RGS11 expression than the epithelial counterparts. Interestingly, RGS11 levels reversibly modulated the epithelial-mesenchymal transition (EMT) of human PDAC cell lines influencing the chemotherapeutic sensitivities of anti-PDAC drugs. Additionally, stable lentiviral-mediated RGS11 expression reduced the cellular proliferation and colony establishment, increased the apoptotic index, and decreased the migratory and invasive abilities in quasi-mesenchymal tumor cell lines, whereas RGS11 depletion in epithelial tumor cell lines showed opposite effects. Global transcriptomic analysis revealed RGS11 replenishment in PDAC cells to suppress CD44-directed stemness features with significant reprogramming of the PDAC oncogenic landscape. Furthermore, RGS11 reduced the primary tumor burden and metastatic occurrence in a mouse model of PDAC. Together, these findings uncover RGS11 as a key target of CNT1 that exhibits therapeutic potential for intervention of aggressive PDAC. Implications: RGS11 identified as a downstream target of a gemcitabine transporter CNT1 exerts potent anti-tumorigenic features in pancreatic ductal adenocarcinoma with therapeutic and prognostic values.

胰腺导管腺癌(PDAC)是一种致命的恶性肿瘤,与早期转移、耐药和预后差有关。我们之前证明了PDAC中浓缩核苷转运蛋白1 (CNT1)的推定肿瘤抑制作用。在这里,我们证明G蛋白信号(RGS) 11的调节因子是CNT1的一个关键靶点,在PDAC中具有有效的肿瘤抑制特性。与正常人胰腺相比,RGS11在人PDAC组织中的表达减少,这与患者生存时间缩短相对应。此外,具有加速生长、转移倾向和对核苷类似物先天抗性的准间充质胰腺肿瘤细胞系的RGS11表达相对较低。有趣的是,RGS11水平可逆地调节人PDAC细胞系的上皮-间质转化(EMT),从而影响抗PDAC药物的化疗敏感性。在准间质肿瘤细胞系中,稳定的慢病毒介导的RGS11表达降低了细胞增殖和集落建立,增加了凋亡指数,降低了迁移和侵袭能力,而在上皮性肿瘤细胞系中,RGS11的表达则表现出相反的作用。全球转录组学分析显示,PDAC细胞中RGS11的补充抑制了cd44导向的干性特征,并显著重编程了PDAC致癌景观。此外,RGS11降低了PDAC小鼠模型的原发肿瘤负荷和转移发生率。总之,这些发现揭示了RGS11作为CNT1的关键靶点,在干预侵袭性PDAC方面表现出治疗潜力。结论:RGS11被确定为吉西他滨转运体CNT1的下游靶点,在胰腺导管腺癌中具有有效的抗肿瘤特性,具有治疗和预后价值。
{"title":"Loss of Regulator of G protein signaling 11 promotes pro-tumorigenic features in pancreatic cancer.","authors":"Tejinder Kaur, Debasis Nayak, Arnav Joshi, Junan Li, Amy Hite, Rajgopal Govindarajan","doi":"10.1158/1541-7786.MCR-25-0144","DOIUrl":"10.1158/1541-7786.MCR-25-0144","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy, associated with early metastasis, drug resistance and poor outcomes. We previously demonstrated a putative tumor suppressive role for concentrative nucleoside transporter 1 (CNT1) in PDAC. Here we demonstrate the regulator of G protein signaling (RGS) 11 as a key target of CNT1, with potent tumor suppressive properties in PDAC. Compared to normal human pancreas, RGS11 expression is diminished in human PDAC tissues which correspond with the reduced patient survival times. In addition, quasi-mesenchymal pancreatic tumor cell lines with accelerated growth, metastatic propensity, and innate resistance to nucleoside analogs showed relatively lower RGS11 expression than the epithelial counterparts. Interestingly, RGS11 levels reversibly modulated the epithelial-mesenchymal transition (EMT) of human PDAC cell lines influencing the chemotherapeutic sensitivities of anti-PDAC drugs. Additionally, stable lentiviral-mediated RGS11 expression reduced the cellular proliferation and colony establishment, increased the apoptotic index, and decreased the migratory and invasive abilities in quasi-mesenchymal tumor cell lines, whereas RGS11 depletion in epithelial tumor cell lines showed opposite effects. Global transcriptomic analysis revealed RGS11 replenishment in PDAC cells to suppress CD44-directed stemness features with significant reprogramming of the PDAC oncogenic landscape. Furthermore, RGS11 reduced the primary tumor burden and metastatic occurrence in a mouse model of PDAC. Together, these findings uncover RGS11 as a key target of CNT1 that exhibits therapeutic potential for intervention of aggressive PDAC. Implications: RGS11 identified as a downstream target of a gemcitabine transporter CNT1 exerts potent anti-tumorigenic features in pancreatic ductal adenocarcinoma with therapeutic and prognostic values.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12680025/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145286250","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
Combined MYC Activation and PTEN Loss Drives Molecular Features of Aggressive Preinvasive Lesions in Mouse Prostate. MYC激活和PTEN缺失联合驱动小鼠前列腺侵袭性侵袭前病变的分子特征。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-13 DOI: 10.1158/1541-7786.MCR-24-1206
Michael Rubenstein, Apurv Rege, Gretchen Hubbard, Danielle Cannady, Shreya Agarwal, Kevin Chen, Alex Estrada, Carolina Gomes Alexandre, Jessica Hicks, Tracy Jones, Qizhi Zheng, Srinivasan Yegnasubramanian, Charles J Bieberich, Angelo M De Marzo

Prostate cancer ranges from indolent to rapidly progressive. An elevated cell proliferation index portends poor outcomes, yet the molecular alterations essential for increased cell proliferation remain ill-defined. Gain of MYC combined with biallelic PTEN loss predicts prostate cancer mortality. Prior studies have shown that combined MYC overexpression and Pten loss, driven by the Hoxb13 locus, results in prostatic intraepithelial neoplastic (PIN) lesions that progress to metastatic disease (BMPC mice). Yet, single gene alterations in these mice result only in PIN. Herein, we performed transcriptomic profiling of PIN lesions from each of the 3 genotypes. While MYC alone resulted in increases in genes related to cell cycle regulation/cell division, combined MYC and Pten loss led to a further and more consistent increase, and a synergistic cell cycle progression. Increased ribosome biogenesis/translation are required for cell proliferation. While MYC alone increased 45S rRNA, and most components of the translation machinery, these were more strongly induced in BMPC mice. Surprisingly, Pten loss alone resulted in a downregulation of translation machinery genes, which could explain the absence of biallelic PTEN loss in human PIN and early carcinomas. Some MYC targets were increased only after Pten loss, indicating Pten loss increases MYC activity. Implications: These findings are that increased cell cycle and translational machinery gene induction may explain the synergy between MYC and PTEN loss for increasing prostate cancer cell proliferation and disease aggressiveness. Finally, these results provide further support for the therapeutic targeting of translation in prostate cancer.

前列腺癌的范围从惰性到快速进展。升高的细胞增殖指数预示着不良的结果,然而增加细胞增殖所必需的分子改变仍然不明确。MYC的增加与双等位基因PTEN的缺失可以预测前列腺癌的死亡率。先前的研究表明,由Hoxb13基因座驱动的MYC过表达和Pten缺失联合导致前列腺上皮内肿瘤(PIN)病变进展为转移性疾病(BMPC小鼠)。然而,这些小鼠的单基因改变只导致PIN。在此,我们对3种基因型的PIN病变进行了转录组学分析。虽然MYC单独导致与细胞周期调节/细胞分裂相关的基因增加,但MYC和Pten的联合缺失导致进一步和更一致的增加,以及协同的细胞周期进展。增加核糖体的生物发生/翻译是细胞增殖所必需的。虽然MYC单独增加了45S rRNA和翻译机制的大多数组成部分,但这些在BMPC小鼠中被更强烈地诱导。令人惊讶的是,Pten缺失单独导致翻译机械基因的下调,这可以解释人类PIN和早期癌症中双等位基因Pten缺失的缺失。一些MYC靶点仅在Pten缺失后增加,表明Pten缺失增加了MYC活性。意义:这些发现表明细胞周期和翻译机制基因诱导的增加可以解释MYC和PTEN缺失之间的协同作用,从而增加前列腺癌细胞增殖和疾病侵袭性。最后,这些结果为翻译靶向治疗前列腺癌提供了进一步的支持。
{"title":"Combined MYC Activation and PTEN Loss Drives Molecular Features of Aggressive Preinvasive Lesions in Mouse Prostate.","authors":"Michael Rubenstein, Apurv Rege, Gretchen Hubbard, Danielle Cannady, Shreya Agarwal, Kevin Chen, Alex Estrada, Carolina Gomes Alexandre, Jessica Hicks, Tracy Jones, Qizhi Zheng, Srinivasan Yegnasubramanian, Charles J Bieberich, Angelo M De Marzo","doi":"10.1158/1541-7786.MCR-24-1206","DOIUrl":"https://doi.org/10.1158/1541-7786.MCR-24-1206","url":null,"abstract":"<p><p>Prostate cancer ranges from indolent to rapidly progressive. An elevated cell proliferation index portends poor outcomes, yet the molecular alterations essential for increased cell proliferation remain ill-defined. Gain of MYC combined with biallelic PTEN loss predicts prostate cancer mortality. Prior studies have shown that combined MYC overexpression and Pten loss, driven by the Hoxb13 locus, results in prostatic intraepithelial neoplastic (PIN) lesions that progress to metastatic disease (BMPC mice). Yet, single gene alterations in these mice result only in PIN. Herein, we performed transcriptomic profiling of PIN lesions from each of the 3 genotypes. While MYC alone resulted in increases in genes related to cell cycle regulation/cell division, combined MYC and Pten loss led to a further and more consistent increase, and a synergistic cell cycle progression. Increased ribosome biogenesis/translation are required for cell proliferation. While MYC alone increased 45S rRNA, and most components of the translation machinery, these were more strongly induced in BMPC mice. Surprisingly, Pten loss alone resulted in a downregulation of translation machinery genes, which could explain the absence of biallelic PTEN loss in human PIN and early carcinomas. Some MYC targets were increased only after Pten loss, indicating Pten loss increases MYC activity. Implications: These findings are that increased cell cycle and translational machinery gene induction may explain the synergy between MYC and PTEN loss for increasing prostate cancer cell proliferation and disease aggressiveness. Finally, these results provide further support for the therapeutic targeting of translation in prostate cancer.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-10-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145280905","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Spatial-in-Spatial: The Utility of Combining Fluorescence-Guided Multiple Sampling with Spatial-Omics in Human Glioblastoma. 空间中的空间:荧光引导多重采样与空间组学相结合在人类胶质母细胞瘤中的应用。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-02 DOI: 10.1158/1541-7786.MCR-25-0194
Shubhang Bhalla, Bethsabe Romero, Yusor Al-Nuaimy, Felix Toussaint, Sina Zoghi, Niels Pacheco-Barrios, Stefan T Prvulovic, Christian A Bowers, Sara G M Piccirillo

Human glioblastoma (GBM) is a remarkable example of a highly aggressive and untreatable tumor. A formidable challenge in treating GBM is its extensive intratumor heterogeneity, which traditional bulk tissue analysis fails to capture. Fluorescence-guided multiple sampling, utilizing 5-aminolevulinic acid for tumor visualization, offers objective tumor tissue identification and enhanced spatial resolution. In this study, we present a perspective on a novel "spatial-in-spatial" approach that enables comprehensive analysis of tumor areas and their microenvironment-at macroscopic and microscopic levels-by combining fluorescence-guided multiple sampling with spatial-omics technologies. This perspective discusses how this integrated methodology has the potential to advance our understanding of GBM biology through the high-resolution, multidimensional characterization of tumor heterogeneity and identification of novel, area-specific therapeutic targets.

人类胶质母细胞瘤是一种高度侵袭性且无法治愈的肿瘤。治疗胶质母细胞瘤的一个巨大挑战是其广泛的肿瘤内异质性,传统的大块组织分析无法捕获。荧光引导多重采样,利用5-氨基乙酰丙酸进行肿瘤可视化,提供客观的肿瘤组织识别和增强的空间分辨率。在这里,我们提出了一种新的“空间中空间”方法的观点,通过将荧光引导的多次采样与空间组学技术相结合,可以在宏观和微观水平上对肿瘤区域及其微环境进行全面分析。这一观点讨论了这种综合方法如何通过高分辨率、多维度表征肿瘤异质性和识别新的、区域特异性治疗靶点来促进我们对胶质母细胞瘤生物学的理解。
{"title":"Spatial-in-Spatial: The Utility of Combining Fluorescence-Guided Multiple Sampling with Spatial-Omics in Human Glioblastoma.","authors":"Shubhang Bhalla, Bethsabe Romero, Yusor Al-Nuaimy, Felix Toussaint, Sina Zoghi, Niels Pacheco-Barrios, Stefan T Prvulovic, Christian A Bowers, Sara G M Piccirillo","doi":"10.1158/1541-7786.MCR-25-0194","DOIUrl":"10.1158/1541-7786.MCR-25-0194","url":null,"abstract":"<p><p>Human glioblastoma (GBM) is a remarkable example of a highly aggressive and untreatable tumor. A formidable challenge in treating GBM is its extensive intratumor heterogeneity, which traditional bulk tissue analysis fails to capture. Fluorescence-guided multiple sampling, utilizing 5-aminolevulinic acid for tumor visualization, offers objective tumor tissue identification and enhanced spatial resolution. In this study, we present a perspective on a novel \"spatial-in-spatial\" approach that enables comprehensive analysis of tumor areas and their microenvironment-at macroscopic and microscopic levels-by combining fluorescence-guided multiple sampling with spatial-omics technologies. This perspective discusses how this integrated methodology has the potential to advance our understanding of GBM biology through the high-resolution, multidimensional characterization of tumor heterogeneity and identification of novel, area-specific therapeutic targets.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"839-843"},"PeriodicalIF":4.7,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144708313","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
PAD2-Mediated Histone Citrullination Drives Tumor Progression by Enhancing Cell Proliferation and Modifying the Microenvironment in Pancreatic Cancer. pad2介导的组蛋白瓜氨酸化通过促进细胞增殖和改变胰腺癌微环境驱动肿瘤进展。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-02 DOI: 10.1158/1541-7786.MCR-24-1095
Kentaro Umemura, Yoshimitsu Akiyama, Shu Shimada, Megumi Hatano, Ayumi Kono, Koya Yasukawa, Atsushi Kamachi, Yosuke Igarashi, Shu Tsukihara, Yoshiaki Tanji, Koichiro Morimoto, Atsushi Nara, Masahiro Yamane, Keiichi Akahoshi, Hiroaki Ono, Akira Shimizu, Yuji Soejima, Minoru Tanabe, Daisuke Ban, Shinji Tanaka

Histone citrullination is catalyzed by peptidyl-arginine deiminases (PAD) that play a role in gene regulation, and several specific inhibitors have been developed. However, the clinical significance, molecular mechanisms of histone citrullination and PADs, and effects of PAD inhibitors in pancreatic ductal adenocarcinoma (PDAC) remain unclear. This study aimed to investigate the role and potential molecular mechanisms of PADs in PDAC. Histone citrullination was upregulated and strongly associated with the nuclear expression of PAD2, one of the PAD family, in human PDAC tissues, correlating with aggressiveness and poor prognosis. PAD2 overexpression increased PDAC cell proliferation, whereas its knockdown had the opposite effect in vitro. PAD2 was recruited to the promoter regions of PRUNE1 and E2F1, resulting in the activation of their mRNA expression via increased histone citrullination and chromatin accessibility. PAD2 overexpression enhanced tumorigenicity and increased PRUNE1 expression and M2 tumor-associated macrophage (M2 TAM) infiltration in vivo. PAD2 inhibitor suppressed the growth and tumorigenicity of PAD2-expressing PDAC mouse models by reducing PRUNE1 expression and M2 macrophage infiltration. Pad2 knockdown and PAD inhibitor treatment showed similar effects in syngeneic mouse models. The triple-high expression of nuclear PAD2, PRUNE1, and the M2 TAM marker CD206 may serve as independent adverse prognostic factors for human PDAC. Conclusively, PAD2-mediated histone citrullination drives PDAC progression by epigenetically regulating downstream target genes and influencing the tumor microenvironment. The PAD2-PRUNE1-M2 TAM axis presents a promising therapeutic target and prognostic indicator for PDAC.

Implications: Elevated PAD2 expression promotes PDAC progression by epigenetically activating PRUNE1 and enhancing M2 macrophage polarization.

组蛋白瓜氨酸化是由参与基因调控的肽基精氨酸脱亚胺酶(PAD)催化的,目前已开发出几种特异性抑制剂。然而,组蛋白瓜氨酸化和PAD的临床意义、分子机制以及PAD抑制剂在胰腺导管腺癌(PDAC)中的作用尚不清楚。本研究旨在探讨pad在PDAC中的作用及其可能的分子机制。在人PDAC组织中,组蛋白瓜氨酸化上调并与PAD家族成员PAD2的核表达密切相关,与侵袭性和不良预后相关。PAD2过表达增加了PDAC细胞的增殖,而其敲低在体外具有相反的作用。PAD2被招募到PRUNE1和E2F1的启动子区域,通过增加组蛋白瓜氨酸化和染色质可及性,激活它们的mRNA表达。PAD2过表达增强了体内的致瘤性,增加了PRUNE1的表达和M2肿瘤相关巨噬细胞(M2 TAM)的浸润。PAD2抑制剂通过降低PRUNE1表达和M2巨噬细胞浸润,抑制表达PAD2的PDAC小鼠模型的生长和致瘤性。在同基因小鼠模型中,Pad2敲除和PAD抑制剂治疗显示出相似的效果。核PAD2、PRUNE1和M2 TAM标记物CD206的三高表达可能是人类PDAC的独立不良预后因素。总之,pad2介导的组蛋白瓜氨酸化通过表观遗传调控下游靶基因和影响肿瘤微环境来驱动PDAC的进展。PAD2-PRUNE1-M2 TAM轴是PDAC的一个有希望的治疗靶点和预后指标。结论:PAD2表达升高通过表观遗传激活PRUNE1和增强M2巨噬细胞极化促进PDAC进展。
{"title":"PAD2-Mediated Histone Citrullination Drives Tumor Progression by Enhancing Cell Proliferation and Modifying the Microenvironment in Pancreatic Cancer.","authors":"Kentaro Umemura, Yoshimitsu Akiyama, Shu Shimada, Megumi Hatano, Ayumi Kono, Koya Yasukawa, Atsushi Kamachi, Yosuke Igarashi, Shu Tsukihara, Yoshiaki Tanji, Koichiro Morimoto, Atsushi Nara, Masahiro Yamane, Keiichi Akahoshi, Hiroaki Ono, Akira Shimizu, Yuji Soejima, Minoru Tanabe, Daisuke Ban, Shinji Tanaka","doi":"10.1158/1541-7786.MCR-24-1095","DOIUrl":"10.1158/1541-7786.MCR-24-1095","url":null,"abstract":"<p><p>Histone citrullination is catalyzed by peptidyl-arginine deiminases (PAD) that play a role in gene regulation, and several specific inhibitors have been developed. However, the clinical significance, molecular mechanisms of histone citrullination and PADs, and effects of PAD inhibitors in pancreatic ductal adenocarcinoma (PDAC) remain unclear. This study aimed to investigate the role and potential molecular mechanisms of PADs in PDAC. Histone citrullination was upregulated and strongly associated with the nuclear expression of PAD2, one of the PAD family, in human PDAC tissues, correlating with aggressiveness and poor prognosis. PAD2 overexpression increased PDAC cell proliferation, whereas its knockdown had the opposite effect in vitro. PAD2 was recruited to the promoter regions of PRUNE1 and E2F1, resulting in the activation of their mRNA expression via increased histone citrullination and chromatin accessibility. PAD2 overexpression enhanced tumorigenicity and increased PRUNE1 expression and M2 tumor-associated macrophage (M2 TAM) infiltration in vivo. PAD2 inhibitor suppressed the growth and tumorigenicity of PAD2-expressing PDAC mouse models by reducing PRUNE1 expression and M2 macrophage infiltration. Pad2 knockdown and PAD inhibitor treatment showed similar effects in syngeneic mouse models. The triple-high expression of nuclear PAD2, PRUNE1, and the M2 TAM marker CD206 may serve as independent adverse prognostic factors for human PDAC. Conclusively, PAD2-mediated histone citrullination drives PDAC progression by epigenetically regulating downstream target genes and influencing the tumor microenvironment. The PAD2-PRUNE1-M2 TAM axis presents a promising therapeutic target and prognostic indicator for PDAC.</p><p><strong>Implications: </strong>Elevated PAD2 expression promotes PDAC progression by epigenetically activating PRUNE1 and enhancing M2 macrophage polarization.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"844-858"},"PeriodicalIF":4.7,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144285772","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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 Novel Bispecific Integrin α5β1/αv Antibody Reprograms the Myc-Regulated Basal Phenotype of Prostate Cancer with NK Cell-Mediated Tumor Elimination. 一种新的双特异性整合素α5β1/αv抗体通过自然杀伤细胞介导的肿瘤消除,重编程myc调控的前列腺癌基础表型。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-02 DOI: 10.1158/1541-7786.MCR-25-0104
Raghav Joshi, Ming Zhou, Jeffrey H Lin, Fei Song, Daniel Fein, Colm Morrissey, Kun Hu, Alexander Poltorak, Paul Mathew

Integrin α5β1 and αv cross-talk in chemotaxis, and clonogenic survival of prostate cancer cells is abrogated by a bispecific α5β1/αv antibody (BsAbα5β1/αv), which uniquely induces internalization and lysosomal degradation of target integrins. We hypothesized that the BsAbα5β1/αv inactivates pathologic mechanosignaling pathways that correlate with integrin expression from patient samples. Mechanistic studies indicate that the BsAbα5β1/αv uniquely reverses Yes-associated protein, β-catenin, and focal adhesion kinase nuclear localization compared with monospecific integrin α5β1 and αv antibody controls in basal-type androgen receptor-negative prostate cancer cells. Dual integrin αv and α5 knockdown alone phenocopied the BsAbα5β1/αv effect. Following BsAbα5β1/αv treatment, Assay for Transposase-Accessible Chromatin using sequencing studies indicated the chromatin accessibility to TEAD and AP-1 family members was markedly reduced. In vitro and in vivo RNA sequencing indicated downregulation of Myc/E2F, TGF-β, and epithelial-mesenchymal transition and upregulation of type I and II IFN transcriptomic pathways. The BsAbα5β1/αv induced CXCL10 and CCL5 cytokine secretion, immune-infiltration of tumors, and NK cell-mediated elimination of the basal-type prostate cancer xenografts in nude mice. αv integrin was highly expressed and principally correlated with the Myc signaling pathway in rapid autopsy tissue microarrays, consistent with correlative data from the SU2C metastatic castration-resistant prostate cancer and Deutsches Krebsforschungszentrum early-onset prostate cancer cohorts. These studies connect integrin signaling with the central biology of basal-type and castration-resistant prostate cancers and define a novel therapeutic strategy that controls critical immunosuppressive pathways.

Implications: Dual integrin α5β1/αv targeting with a bispecific antibody represents a novel therapeutic strategy that reprograms the epigenetic and transcriptomic signatures of basal-type prostate cancer with induction of immunologic tumor control.

一种双特异性α5β1/αv抗体(BsAbα5β1/αv)消除了整合素α5β1和αv在前列腺癌细胞趋化性和克隆存活中的串扰,该抗体独特地诱导目标整合素的内化和溶酶体降解。我们假设BsAbα5β1/αv灭活了与患者样本中整合素表达相关的病理机械信号通路。机制研究表明,与单特异性整合素α5β1和αv抗体对照相比,BsAbα5β1/αv在基底型雄激素受体阴性前列腺癌细胞中独特地逆转YAP、β -catenin和FAK的核定位。双整联素αv和α5敲低均表现了BsAbα5β1/αv效应。在BsAbα5β1/αv处理后,ATAC-seq研究表明,TEAD和AP-1家族成员的染色质可及性明显降低。体外和体内RNA-seq显示Myc/E2F、tgf - β和上皮间充质转化(epithelial mesenchymal transition, EMT)下调,I型和II型干扰素转录组通路上调。BsAbα5β1/αv诱导CXCL10和CCL5细胞因子分泌、肿瘤免疫浸润和自然杀伤细胞介导的基底型前列腺癌异种移植裸鼠的消除。在快速尸检组织微阵列中,αv整合素高表达并主要与Myc信号通路相关,这与SU2C转移性去势抵抗性前列腺癌和DKFZ早发性前列腺癌队列的相关数据一致。这些研究将整合素信号与基底型和去势抵抗性前列腺癌的中心生物学联系起来,并确定了一种控制关键免疫抑制途径的新治疗策略。意义:双特异性抗体靶向双整合素α5β1/αv代表了一种新的治疗策略,通过诱导免疫肿瘤控制来重编程基底型前列腺癌的表观遗传和转录组特征。
{"title":"A Novel Bispecific Integrin α5β1/αv Antibody Reprograms the Myc-Regulated Basal Phenotype of Prostate Cancer with NK Cell-Mediated Tumor Elimination.","authors":"Raghav Joshi, Ming Zhou, Jeffrey H Lin, Fei Song, Daniel Fein, Colm Morrissey, Kun Hu, Alexander Poltorak, Paul Mathew","doi":"10.1158/1541-7786.MCR-25-0104","DOIUrl":"10.1158/1541-7786.MCR-25-0104","url":null,"abstract":"<p><p>Integrin α5β1 and αv cross-talk in chemotaxis, and clonogenic survival of prostate cancer cells is abrogated by a bispecific α5β1/αv antibody (BsAbα5β1/αv), which uniquely induces internalization and lysosomal degradation of target integrins. We hypothesized that the BsAbα5β1/αv inactivates pathologic mechanosignaling pathways that correlate with integrin expression from patient samples. Mechanistic studies indicate that the BsAbα5β1/αv uniquely reverses Yes-associated protein, β-catenin, and focal adhesion kinase nuclear localization compared with monospecific integrin α5β1 and αv antibody controls in basal-type androgen receptor-negative prostate cancer cells. Dual integrin αv and α5 knockdown alone phenocopied the BsAbα5β1/αv effect. Following BsAbα5β1/αv treatment, Assay for Transposase-Accessible Chromatin using sequencing studies indicated the chromatin accessibility to TEAD and AP-1 family members was markedly reduced. In vitro and in vivo RNA sequencing indicated downregulation of Myc/E2F, TGF-β, and epithelial-mesenchymal transition and upregulation of type I and II IFN transcriptomic pathways. The BsAbα5β1/αv induced CXCL10 and CCL5 cytokine secretion, immune-infiltration of tumors, and NK cell-mediated elimination of the basal-type prostate cancer xenografts in nude mice. αv integrin was highly expressed and principally correlated with the Myc signaling pathway in rapid autopsy tissue microarrays, consistent with correlative data from the SU2C metastatic castration-resistant prostate cancer and Deutsches Krebsforschungszentrum early-onset prostate cancer cohorts. These studies connect integrin signaling with the central biology of basal-type and castration-resistant prostate cancers and define a novel therapeutic strategy that controls critical immunosuppressive pathways.</p><p><strong>Implications: </strong>Dual integrin α5β1/αv targeting with a bispecific antibody represents a novel therapeutic strategy that reprograms the epigenetic and transcriptomic signatures of basal-type prostate cancer with induction of immunologic tumor control.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"873-888"},"PeriodicalIF":4.7,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12323549/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144476115","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
LINC01235 Is an Upstream Regulator of the NFIB Gene and the Notch Pathway in Triple-Negative Breast Cancer. LINC01235是三阴性乳腺癌中NFIB基因和NOTCH通路的上游调控因子。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-02 DOI: 10.1158/1541-7786.MCR-24-1143
Wenbo Xu, Sonam Bhatia, Yunus Sahin, David L Spector

We identified a long noncoding RNA, LINC01235, with significant enrichment in luminal progenitor (LP)-like cells in triple-negative breast cancer (TNBC) organoids and cell lines. Antisense-mediated knockdown or genetic knockout of LINC01235 in TNBC cell lines led to a decline in cell proliferation and adversely affected the ability to form organoids. A comprehensive co-expression analysis, leveraging The Cancer Genome Atlas data, revealed a distinct correlation between LINC01235 expression and the expression of NFIB, a neighboring gene encoding a transcription factor. Subsequent CRISPR knockout or antisense oligonucleotide-mediated knockdown studies demonstrated an upstream regulatory role of LINC01235 over NFIB. Moreover, our investigations demonstrated that LINC01235 regulates the Notch pathway through NFIB, and chromatin isolation by RNA purification followed by qPCR results indicated the direct binding of LINC01235 to the NFIB promoter. Our findings demonstrate that LINC01235 positively regulates NFIB transcription, which in turn modulates the Notch pathway, influencing LP-like cell proliferation in breast cancer progression. This study highlights a pivotal role of LINC01235 in TNBC and its potential as a therapeutic target.

Implications: This study demonstrates the central role of LINC01235 as an upstream positive regulator of NFIB and the Notch signaling pathway to induce the production of LP-like cells in TNBC.

我们发现了一个长链非编码RNA (lncRNA), LINC01235,在三阴性乳腺癌类器官和细胞系的腔祖(LP)样细胞中显著富集。在TNBC细胞系中,反义介导的LINC01235敲低或基因敲除导致细胞增殖下降,并对形成类器官的能力产生不利影响。利用TCGA数据进行的综合共表达分析显示,LINC01235的表达与编码转录因子的邻近基因NFIB的表达之间存在明显的相关性。随后的CRISPR敲除或aso介导的敲除研究证实了LINC01235对NFIB的上游调控作用。此外,我们的研究表明,LINC01235通过NFIB调控NOTCH通路,ChIRP-qPCR结果表明LINC01235与NFIB启动子直接结合。我们的研究结果表明,LINC01235正调控NFIB转录,进而调节NOTCH通路,影响lp样细胞在乳腺癌进展中的增殖。这项研究强调了LINC01235在TNBC中的关键作用及其作为治疗靶点的潜力。本研究表明,LINC01235作为NFIB和NOTCH信号通路的上游正调节因子,在TNBC中诱导管腔祖样细胞的产生中起着核心作用。
{"title":"LINC01235 Is an Upstream Regulator of the NFIB Gene and the Notch Pathway in Triple-Negative Breast Cancer.","authors":"Wenbo Xu, Sonam Bhatia, Yunus Sahin, David L Spector","doi":"10.1158/1541-7786.MCR-24-1143","DOIUrl":"10.1158/1541-7786.MCR-24-1143","url":null,"abstract":"<p><p>We identified a long noncoding RNA, LINC01235, with significant enrichment in luminal progenitor (LP)-like cells in triple-negative breast cancer (TNBC) organoids and cell lines. Antisense-mediated knockdown or genetic knockout of LINC01235 in TNBC cell lines led to a decline in cell proliferation and adversely affected the ability to form organoids. A comprehensive co-expression analysis, leveraging The Cancer Genome Atlas data, revealed a distinct correlation between LINC01235 expression and the expression of NFIB, a neighboring gene encoding a transcription factor. Subsequent CRISPR knockout or antisense oligonucleotide-mediated knockdown studies demonstrated an upstream regulatory role of LINC01235 over NFIB. Moreover, our investigations demonstrated that LINC01235 regulates the Notch pathway through NFIB, and chromatin isolation by RNA purification followed by qPCR results indicated the direct binding of LINC01235 to the NFIB promoter. Our findings demonstrate that LINC01235 positively regulates NFIB transcription, which in turn modulates the Notch pathway, influencing LP-like cell proliferation in breast cancer progression. This study highlights a pivotal role of LINC01235 in TNBC and its potential as a therapeutic target.</p><p><strong>Implications: </strong>This study demonstrates the central role of LINC01235 as an upstream positive regulator of NFIB and the Notch signaling pathway to induce the production of LP-like cells in TNBC.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"859-872"},"PeriodicalIF":4.7,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12221212/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528958","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
Myeloid-Derived Suppressor Cells: Orchestrators of Tumor Immune Evasion and Therapeutic Vulnerabilities. 髓源性抑制细胞:肿瘤免疫逃避和治疗脆弱性的协调者。
IF 4.7 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-10-02 DOI: 10.1158/1541-7786.MCR-25-0251
Ziyu Wang, Xiaoping Du, Xiangxue Xing, Wenjing Xie, Haina Xin, Wan Liu

Myeloid-derived suppressor cells (MDSCs) are characterized by abnormal phenotypes, high heterogeneity, and immunosuppressive function. MDSCs are critical components in the tumor immune microenvironment, contributing to cancer progression by inhibiting T cells, B cells, NK cells, and dendritic cells while promoting regulatory T cells, tumor-associated macrophages, and Th17 cells. Beyond immunosuppression, MDSCs facilitate tumor angiogenesis, tumor cell stemness, epithelial-mesenchymal transition, and premetastatic niche formation. Current therapeutic strategies targeting MDSCs include depletion, functional inhibition, induction of differentiation, and disruption of MDSC recruitment and activation. Various therapeutic agents-including chemotherapeutics, mAbs, small-molecule inhibitors, and natural compounds-have shown efficacy in modulating MDSC activity. Combining MDSC-targeted therapy with existing immunotherapies, such as immune checkpoint inhibitors, may further improve antitumor responses.

髓源性抑制细胞(MDSCs)具有异常表型、高异质性和免疫抑制功能。MDSCs是肿瘤免疫微环境的关键组成部分,通过抑制T细胞、B细胞、自然杀伤细胞和树突状细胞,同时促进调节性T细胞、肿瘤相关巨噬细胞和辅助性T细胞的产生,从而促进癌症的进展。除了免疫抑制外,MDSCs还促进肿瘤血管生成、肿瘤细胞干细胞、上皮-间质转化和转移前生态位的形成。目前针对MDSCs的治疗策略包括耗竭、功能抑制、诱导分化和破坏MDSC的募集和激活。各种治疗药物,包括化疗药物、单克隆抗体、小分子抑制剂和天然化合物,已经显示出调节MDSC活性的功效。结合mdsc靶向治疗与现有的免疫疗法,如免疫检查点抑制剂,可能进一步改善抗肿瘤反应。
{"title":"Myeloid-Derived Suppressor Cells: Orchestrators of Tumor Immune Evasion and Therapeutic Vulnerabilities.","authors":"Ziyu Wang, Xiaoping Du, Xiangxue Xing, Wenjing Xie, Haina Xin, Wan Liu","doi":"10.1158/1541-7786.MCR-25-0251","DOIUrl":"10.1158/1541-7786.MCR-25-0251","url":null,"abstract":"<p><p>Myeloid-derived suppressor cells (MDSCs) are characterized by abnormal phenotypes, high heterogeneity, and immunosuppressive function. MDSCs are critical components in the tumor immune microenvironment, contributing to cancer progression by inhibiting T cells, B cells, NK cells, and dendritic cells while promoting regulatory T cells, tumor-associated macrophages, and Th17 cells. Beyond immunosuppression, MDSCs facilitate tumor angiogenesis, tumor cell stemness, epithelial-mesenchymal transition, and premetastatic niche formation. Current therapeutic strategies targeting MDSCs include depletion, functional inhibition, induction of differentiation, and disruption of MDSC recruitment and activation. Various therapeutic agents-including chemotherapeutics, mAbs, small-molecule inhibitors, and natural compounds-have shown efficacy in modulating MDSC activity. Combining MDSC-targeted therapy with existing immunotherapies, such as immune checkpoint inhibitors, may further improve antitumor responses.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":" ","pages":"829-838"},"PeriodicalIF":4.7,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144775868","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Molecular 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:604180095
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