Epigenetic regulation of pancreatic cancer subtype identity and tumorigenesis by PHF2
PHF2 对胰腺癌亚型识别和肿瘤发生的表观遗传调控
基本信息
- 批准号:10657989
- 负责人:
- 金额:$ 37.89万
- 依托单位:
- 依托单位国家:美国
- 项目类别:
- 财政年份:2023
- 资助国家:美国
- 起止时间:2023-07-07 至 2028-06-30
- 项目状态:未结题
- 来源:
- 关键词:3-DimensionalAcinar CellAcuteArchitectureBiologicalBiologyCRISPR/Cas technologyCSPG6 geneCancer EtiologyCell Culture TechniquesCell LineCellsCessation of lifeChemoresistanceChromatinChromatin LoopChromosomesCo-ImmunoprecipitationsCollectionDNADataDevelopmentDisease ResistanceEpigenetic ProcessEvolutionExpression ProfilingGene ExpressionGenesGenetic ScreeningGenetic TranscriptionGenetically Engineered MouseGrowth and Development functionHuman Cell LineInvestigationKRAS2 geneMaintenanceMalignant NeoplasmsMalignant neoplasm of pancreasMapsMediatingMediatorMesenchymalModelingMolecular TargetMutationOutcomePancreasPancreatic Ductal AdenocarcinomaPatient-Focused OutcomesPatientsProteinsPublishingRegimenResearchResolutionRoleSpecimenSystemTestingTherapeuticTherapeutic InterventionTumor-infiltrating immune cellsUnited StatesWorkcancer invasivenesscancer subtypescell typechemotherapycohesincohesiondefined contributiondigitalepigenetic regulationexperimental studyhistone demethylasehuman RNA sequencingin vivoinnovationinsightmolecular subtypesmouse modelnano-stringnovelnovel therapeutic interventionnovel therapeuticspancreas developmentpancreatic ductal adenocarcinoma cellpancreatic ductal adenocarcinoma modelpancreatic neoplasmpatient responsepatient subsetspre-clinicalprogenitorprogramsrecruitresponsesingle-cell RNA sequencingtherapeutic targettranscriptome sequencingtranscriptomicstumortumor growthtumor initiationtumor microenvironmenttumor xenografttumor-immune system interactionstumorigenesisvirtual
项目摘要
Molecular subtyping of pancreatic ductal adenocarcinoma (PDAC) has defined two major transcriptional
subtypes, Classical and Basal-like, which are presently the most powerful predictors of patient outcomes.
Thus, there is a critical need to determine the underlying mechanisms responsible for PDAC cell identity.
Our preliminary experiments demonstrate that inactivation of PHF2, a histone demethylase, in low-passage
patient-derived Classical PDAC cells results in a striking loss of the Classical gene program and virtually
abolishes tumor growth. In addition, deletion of Phf2 suppresses pancreatic tumor initiation and prolongs
survival in genetically engineered mouse models. Moreover, our CUT&RUN experiments reveal that PHF2
co-localizes on chromosomes with CTCF and SMC3, proteins that contribute to cell identity by regulating
the three-dimensional architecture of chromatin. Based on these exciting preliminary results, our central
hypothesis is that PHF2, controls the Classical gene program in PDAC by regulating three-dimensional
architecture of chromatin. Experiments in this proposal will employ a unique collection of low-passage
human cell lines, high resolution mapping of chromatin-associated proteins and chromatin architecture, and
NanoString GeoMx spatial transcriptomic analysis of genetically engineered mouse models to define the
role of PHF2 in PDAC. Three specific aims are proposed to test the central hypothesis: 1) Define the subsets
of PDAC that are dependent on PHF2; 2) Identify the mechanisms by which PHF2 regulates gene expression
in PDAC; and 3) Define the contribution of Phf2 to tumorigenesis in genetically engineered mice. In the first
aim, orthotopic xenograft tumor models allowing for the temporal inactivation of PHF2 by CRISPR/Cas9 will
be employed to determine the importance of PHF2 in the maintenance of PDAC tumors as well as their
response to chemotherapy. In the second aim, CUT&RUN, HiChIP, and RNA-seq experiments will be
employed to define the global localization of PHF2 on chromatin in PDAC cells and ascertain its role in
recruiting CTCF and cohesion to DNA to mediate DNA loop formation. For the third aim, genetically
engineered mouse models of PDAC lacking Phf2 will be used to elucidate its role in the KRAS-driven
changes in cell identity required for pancreatic acinar cells to transform to invasive cancer . Spatial
transcriptomic studies will be performed to ascertain how Phf2 deficiency alters the composition of the tumor
microenvironment by the development of PDAC that is deficient in the Classical program. The research
proposed in this application is innovative because it will define a novel function for PHF2 in regulating
chromatin architecture, further elucidating the mechanisms that allow for cell type-specific chromatin
architecture. The proposed research is significant because it will provide strong evidence for the role of
epigenetic regulation of PDAC cell identity, ultimately providing new opportunities for the development of
novel therapies for PDAC.
胰腺导管腺癌 (PDAC) 的分子分型定义了两个主要的转录
亚型,经典型和基础型,目前是患者结果最有力的预测因子。
因此,迫切需要确定负责 PDAC 细胞身份的潜在机制。
我们的初步实验表明,PHF2(一种组蛋白去甲基化酶)在低传代中失活
患者来源的经典 PDAC 细胞会导致经典基因程序的显着丧失,并且实际上
消除肿瘤生长。此外,Phf2 的缺失可抑制胰腺肿瘤的发生并延长胰腺肿瘤的存活时间。
基因工程小鼠模型中的存活率。此外,我们的 CUT&RUN 实验表明 PHF2
与 CTCF 和 SMC3 共定位在染色体上,这些蛋白质通过调节来促进细胞身份
染色质的三维结构。基于这些令人兴奋的初步结果,我们的中央
假设 PHF2 通过调节三维空间来控制 PDAC 中的经典基因程序
染色质的结构。本提案中的实验将采用独特的低通道集合
人类细胞系、染色质相关蛋白和染色质结构的高分辨率图谱,以及
NanoString GeoMx 对基因工程小鼠模型进行空间转录组分析,以定义
PHF2 在 PDAC 中的作用。提出了三个具体目标来检验中心假设:1)定义子集
依赖于 PHF2 的 PDAC; 2) 确定PHF2调节基因表达的机制
在 PDAC 中; 3) 确定 Phf2 对基因工程小鼠肿瘤发生的贡献。在第一个
目的,允许通过 CRISPR/Cas9 暂时失活 PHF2 的原位异种移植肿瘤模型将
用于确定 PHF2 在 PDAC 肿瘤及其维持中的重要性
对化疗的反应。在第二个目标中,CUT&RUN、HiChIP 和 RNA-seq 实验将
用于定义 PHF2 在 PDAC 细胞染色质上的全局定位并确定其在
募集 CTCF 和 DNA 内聚力来介导 DNA 环的形成。对于第三个目标,从基因上来说
缺乏 Phf2 的 PDAC 工程小鼠模型将用于阐明其在 KRAS 驱动的
胰腺腺泡细胞转变为侵袭性癌症所需的细胞特性的变化。空间
将进行转录组研究以确定 Phf2 缺乏如何改变肿瘤的组成
通过开发 PDAC 来弥补经典程序中存在的缺陷。研究
本申请中提出的方案具有创新性,因为它将定义 PHF2 在调节方面的新功能
染色质结构,进一步阐明细胞类型特异性染色质的机制
建筑学。拟议的研究意义重大,因为它将为证明其作用提供强有力的证据
PDAC细胞身份的表观遗传调控,最终为PDAC细胞的发展提供新的机遇
PDAC 的新疗法。
项目成果
期刊论文数量(0)
专著数量(0)
科研奖励数量(0)
会议论文数量(0)
专利数量(0)
数据更新时间:{{ journalArticles.updateTime }}
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
数据更新时间:{{ journalArticles.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ monograph.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ sciAawards.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ conferencePapers.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ patent.updateTime }}
Andrew Liss其他文献
Andrew Liss的其他文献
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
{{ truncateString('Andrew Liss', 18)}}的其他基金
Epigenetic Regulation of Hemidesmosome Signaling in Pancreatic Cancer
胰腺癌半桥粒信号的表观遗传调控
- 批准号:
10046923 - 财政年份:2020
- 资助金额:
$ 37.89万 - 项目类别:
相似国自然基金
BCL2L1/FUNDC1介导的线粒体自噬失衡调控腺泡细胞铁死亡在急性胰腺炎发病中的作用及机制研究
- 批准号:82370651
- 批准年份:2023
- 资助金额:49 万元
- 项目类别:面上项目
清胰颗粒调控重症急性胰腺炎腺泡细胞脂质代谢重塑机制诠释“通腑泻浊”新内涵
- 批准号:82374248
- 批准年份:2023
- 资助金额:49 万元
- 项目类别:面上项目
衣康酸介导KDM3A/H3K9/PARP9轴调控DNA损伤修复在急性胰腺炎腺泡细胞坏死中的作用和机制研究
- 批准号:82370653
- 批准年份:2023
- 资助金额:49 万元
- 项目类别:面上项目
线粒体丙酮酸载体调控腺泡细胞糖代谢和钙超载减轻急性胰腺炎的功能与机制研究
- 批准号:82270672
- 批准年份:2022
- 资助金额:52 万元
- 项目类别:面上项目
内质网应激通过sXBP1/HIF-1α/mTOR轴调控腺泡细胞缺陷性自噬在急性胰腺炎发病中的作用及其机制
- 批准号:
- 批准年份:2022
- 资助金额:52 万元
- 项目类别:面上项目
相似海外基金
R21 MPI microRNA directed therapy for treating early stage pancreatic cancer
R21 MPI microRNA 定向疗法治疗早期胰腺癌
- 批准号:
10577609 - 财政年份:2023
- 资助金额:
$ 37.89万 - 项目类别:
Determining Sox10-mediated plasticity in irradiated salivary gland cells
确定受辐射唾液腺细胞中 Sox10 介导的可塑性
- 批准号:
10606665 - 财政年份:2023
- 资助金额:
$ 37.89万 - 项目类别:
Elucidating the role of Fra1 in pancreatic Kras-driven acinar to ductal metaplasia
阐明 Fra1 在胰腺 Kras 驱动的腺泡到导管化生中的作用
- 批准号:
10537870 - 财政年份:2022
- 资助金额:
$ 37.89万 - 项目类别:
Elucidating the role of Fra1 in pancreatic Kras-driven acinar to ductal metaplasia
阐明 Fra1 在胰腺 Kras 驱动的腺泡到导管化生中的作用
- 批准号:
10631947 - 财政年份:2022
- 资助金额:
$ 37.89万 - 项目类别:
Epithelial-immune cell crosstalk during injury and recovery in acute pancreatitis
急性胰腺炎损伤和恢复过程中上皮-免疫细胞的串扰
- 批准号:
10543109 - 财政年份:2021
- 资助金额:
$ 37.89万 - 项目类别: