Gut Microbes’ Impact on Oncogenic Drivers: Location Matters

生物 突变体 癌变 癌基因 癌症研究 微生物群 癌症 抑制器 肠道菌群 斯科普斯 遗传学 免疫学 细胞周期 基因 生物化学 梅德林
作者
Michael G. White,Jennifer A. Wargo
出处
期刊:Molecular Cell [Elsevier]
卷期号:79 (6): 878-880 被引量:2
标识
DOI:10.1016/j.molcel.2020.08.021
摘要

Recent work by Kadosh et al., 2020Kadosh E. Snir-Alkalay I. Venkatachalam A. May S. Lasry A. Elyada E. Zinger A. Shaham M. Vaalani G. Mernberger M. et al.The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic.Nature. 2020; (Published online July 29, 2020)https://doi.org/10.1038/s41586-020-2541-0Crossref Scopus (50) Google Scholar suggests that mutant p53 activity in gut epithelia is influenced by local production of microbial metabolites. The switch of p53 from tumor suppressor to oncogene is location-dependent and is impacted by microbially derived gallic acid. Recent work by Kadosh et al., 2020Kadosh E. Snir-Alkalay I. Venkatachalam A. May S. Lasry A. Elyada E. Zinger A. Shaham M. Vaalani G. Mernberger M. et al.The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic.Nature. 2020; (Published online July 29, 2020)https://doi.org/10.1038/s41586-020-2541-0Crossref Scopus (50) Google Scholar suggests that mutant p53 activity in gut epithelia is influenced by local production of microbial metabolites. The switch of p53 from tumor suppressor to oncogene is location-dependent and is impacted by microbially derived gallic acid. The impact of microbes within tumors and within the gut is increasingly being recognized in the field of cancer—both in their potential contributions to carcinogenesis (Sears and Garrett, 2014Sears C.L. Garrett W.S. Microbes, microbiota, and colon cancer.Cell Host Microbe. 2014; 15: 317-328Abstract Full Text Full Text PDF PubMed Scopus (428) Google Scholar), as well as in modulating therapeutic response (Gopalakrishnan et al., 2018Gopalakrishnan V. Spencer C.N. Nezi L. Reuben A. Andrews M.C. Karpinets T.V. Prieto P.A. Vicente D. Hoffman K. Wei S.C. et al.Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients.Science. 2018; 359: 97-103Crossref PubMed Scopus (1403) Google Scholar; Zitvogel et al., 2017Zitvogel L. Daillère R. Roberti M.P. Routy B. Kroemer G. Anticancer effects of the microbiome and its products.Nat. Rev. Microbiol. 2017; 15: 465-478Crossref PubMed Scopus (183) Google Scholar). The characterization of these microbes has been greatly facilitated via next-generation sequencing (NGS) approaches, with microbial signatures now identified within tumors across cancer types (Poore et al., 2020Poore G.D. Kopylova E. Zhu Q. Carpenter C. Fraraccio S. Wandro S. Kosciolek T. Janssen S. Metcalf J. Song S.J. et al.Microbiome analyses of blood and tissues suggest cancer diagnostic approach.Nature. 2020; 579: 567-574Crossref PubMed Scopus (100) Google Scholar), offering new diagnostic and therapeutic opportunities. Insights are being gained into the mechanisms through which these microbes may positively or negatively impact carcinogenesis and therapeutic response (Bullman et al., 2017Bullman S. Pedamallu C.S. Sicinska E. Clancy T.E. Zhang X. Cai D. Neuberg D. Huang K. Guevara F. Nelson T. et al.Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer.Science. 2017; 358: 1443-1448Crossref PubMed Scopus (404) Google Scholar; Geller et al., 2017Geller L.T. Barzily-Rokni M. Danino T. Jonas O.H. Shental N. Nejman D. Gavert N. Zwang Y. Cooper Z.A. Shee K. et al.Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine.Science. 2017; 357: 1156-1160Crossref PubMed Scopus (384) Google Scholar; Pushalkar et al., 2018Pushalkar S. Hundeyin M. Daley D. Zambirinis C.P. Kurz E. Mishra A. Mohan N. Aykut B. Usyk M. Torres L.E. et al.The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression.Cancer Discov. 2018; 8: 403-416Crossref PubMed Scopus (277) Google Scholar; Riquelme et al., 2019Riquelme E. Zhang Y. Zhang L. Montiel M. Zoltan M. Dong W. Quesada P. Sahin I. Chandra V. San Lucas A. et al.Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes..Cell. 2019; 178: 795-806.e12Abstract Full Text Full Text PDF PubMed Scopus (231) Google Scholar), including associations with oncogenic drivers such as p53 (Greathouse et al., 2018Greathouse K.L. White J.R. Vargas A.J. Bliskovsky V.V. Beck J.A. von Muhlinen N. Polley E.C. Bowman E.D. Khan M.A. Robles A.I. et al.Interaction between the microbiome and TP53 in human lung cancer.Genome Biol. 2018; 19: 123Crossref PubMed Scopus (73) Google Scholar). These mechanisms, however, are incompletely understood and more research is clearly needed. Microbes are known to produce a multitude of metabolites that can impact host physiology and may also impact carcinogenesis as well as therapy response (Zitvogel et al., 2017Zitvogel L. Daillère R. Roberti M.P. Routy B. Kroemer G. Anticancer effects of the microbiome and its products.Nat. Rev. Microbiol. 2017; 15: 465-478Crossref PubMed Scopus (183) Google Scholar). Additionally, microbial diversity and composition differs substantially based on the location within the gut—thus, the local impact of microbially produced metabolites and other microbial mechanisms may be context dependent. A recent manuscript by Kadosh and colleagues published in Nature illustrates this point, demonstrating the impact of microbial metabolites on activity of mutant p53 at different locations within the gut (Kadosh et al., 2020Kadosh E. Snir-Alkalay I. Venkatachalam A. May S. Lasry A. Elyada E. Zinger A. Shaham M. Vaalani G. Mernberger M. et al.The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic.Nature. 2020; (Published online July 29, 2020)https://doi.org/10.1038/s41586-020-2541-0Crossref Scopus (50) Google Scholar). Using murine and organoid models of carcinogenesis, Kadosh and colleagues demonstrated dichotomous activity of mutant p53 as a tumor suppressor versus an oncogene in two different locations within the gut. Importantly, this was influenced by metabolites produced by the local gut microbiota, with divergent activities of mutant p53 in the proximal versus distal gut. This was associated with different levels of the metabolite gallic acid. In these studies, the group used two mouse models to study the impact of mutant p53 on gut tumorigenesis: CKIα mice, in which dysplasia was measured via immunohistochemistry (IHC), and ApcMin/+ mice, in which discrete intestinal tumors were formed. Initially, the authors observed dysplasia in the jejunum and ileum of mice with CKIα and wild-type p53 conditionally deleted from the gut epithelium (CKIαΔgut, p53Δgut), while mice with a p53 R172H mutation (p53R172H) demonstrated dysplasia confined only to the ileum. A similar effect was seen in ApcMin/+ mice whose tumors were primarily in the jejunum with sparing of the colon with wild-type p53 (p53WT) while mutated p53R172H acted as a tumor suppressor in the jejunum but oncogenic in the colon. These effects were then shown to be secondary to dysregulation of the WNT pathway through IHC analysis of WNT target genes. This initial signal demonstrated mutant p53’s action to be spatially dependent in vivo, acting as a tumor suppressor in the proximal gut and as an oncogene in the distal gut. The above spatial relationship, while compelling, does not speak to the source of this observed heterogeneity. Differences in dysplasia by cell type were then studied in ex vivo organoid models of ileal and jejunal cells. The authors noted that p53R172H as well as common mutations in human cancers p53R175H and p53273H exhibited tumor suppressive function in both CKIα and APC organoid models regardless of the sources of those organoids (jejunal versus ileal). This suggests the demonstrated mutated p53 tumor suppressor function regardless of cell type in vitro. In vivo factors, such as the gut microbiota, were then considered the possible causative factor. The group then returned to the in vivo murine model and demonstrated that treatment of mice with antibiotics (with resulting depletion of the gut microbiota confirmed with 16 s sequencing) was associated with restored tumor suppressor function of mutant p53R172H in the distal gut, whereas mice with conditional knockout of mutant p53 continued to demonstrate dysplasia in the proximal and distal gut. It was again confirmed that this tumor suppressive effect was associated with loss of WNT-dependent hyperproliferation. To study more precisely the action of the distal gut microbiome causing this switch, the authors’ attention moved to potentially causative metabolomics in the distal gut. To do this, they studied jejunal CKIαKOp53R172H organoids with a screen for metabolites with known potential impact on tumorogenesis via epigenetic modulation of gene expression. In these studies, the presence of high concentrations of gallic acid was associated with increased proliferative capacity of the organoids—and this was only true in the CKIαKOp53R17H organoids. Notably, hyperproliferative potential of the organoids was lost when gallic acid was removed from the media. The group then brought this back to in vivo models, demonstrating markedly higher levels of gallic acid in the ileum versus the jejunum along with differential expression of the bacterial enzyme shikimate dehydrogenase, which is involved in gallic acid production. Treatment with exogenous gallic acid was associated with highly proliferative epithelia in the jejunum with lesions of high-grade dysplasia in the mutant p53 background (but not with the wild-type p53 background), suggesting that this is mediated through mutant p53. This appeared to be mediated at least in part via gallic acid’s impact on TCF-4-chromatin association and H3K4me3 modification at genomic WNT promoters in jejunal enterocytes harboring mutant p53 (Figure 1). Together, these data galvanize the concept that gut microbes can have a profound impact on carcinogenesis and also highlight the plasticity of cancer-associated genes such as p53 and their influence from the local microenvironment. Second, it highlights that functional aspects of microbial environments are likely to have a dominant role via impact of metabolites and other factors on host physiology in health and disease. Thus, it is critical as we move forward as a field to focus on functional aspects of gut microbiota and their impact, rather than solely focusing on taxonomy. Nonetheless limitations exist with our current understanding, and further research exploring such associations and the mechanisms behind this are critically needed. Research studies in pre-clinical models such as the one performed by Kadosh et al. are important, though the relevance of such findings in patients needs to be carefully understood before bringing forward therapeutic approaches (such as targeting gallic acid). Though such research and reductionist approaches in genetically engineered mouse models are helpful in elucidating potential mechanisms, interactions in human organisms are far more complex with multiple potential confounders. Nonetheless, such studies, coupled with human subjects research, are paramount in developing next generation strategies for cancer care. J.A.W. is supported by the NIH ( 1 R01 CA219896-01A1 ), U.S-Israel Binational Science Foundation ( 201332 ), the Melanoma Research Alliance ( 4022024 ), American Association for Cancer Research Stand Up To Cancer ( SU2C-AACR-IRG-19-17 ), Department of Defense ( W81XWH-16-1-0121 ), MD Anderson Cancer Center Multidisciplinary Research Program Grant , Andrew Sabin Family Fellows Program , and MD Anderson Cancer Center’s Melanoma Moon Shots Program . J.A.W. is a member of the Parker Institute for Cancer Immunotherapy at MD Anderson Cancer Center. J.A.W. is an inventor on a US patent application (PCT/US17/53.717) submitted by the University of Texas MD Anderson Cancer Center that covers methods to enhance immune checkpoint blockade responses by modulating the microbiome. J.A.W. reports compensation for speaker’s bureau and honoraria from Imedex, Dava Oncology, Omniprex, Illumina, Gilead, PeerView, Physician Education Resource, MedImmune, and Bristol-Myers Squibb. J.A.W. serves as a consultant/advisory board member for Roche/Genentech, Novartis, AstraZeneca, GlaxoSmithKline, Bristol-Myers Squibb, Merck, Biothera Pharmaceuticals, and Microbiome DX. J.A.W. also receives research support from GlaxoSmithKline, Roche/Genentech, Bristol-Myers Squibb, and Novartis.

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
1秒前
哈哈哈哈哈完成签到,获得积分10
3秒前
wpy发布了新的文献求助20
4秒前
4秒前
rocky15应助刻苦的惜梦采纳,获得50
7秒前
rediculous发布了新的文献求助10
7秒前
Annie发布了新的文献求助10
7秒前
酷炫的雅琴应助招财小茗采纳,获得20
11秒前
天天快乐应助xiuwenli采纳,获得10
13秒前
rediculous完成签到,获得积分10
14秒前
所所应助欣喜的代容采纳,获得10
14秒前
14秒前
15秒前
18秒前
失眠的哈密瓜完成签到,获得积分10
18秒前
LiuSheng发布了新的文献求助10
19秒前
发糕太甜了应助研友_Z7Xdl8采纳,获得10
21秒前
22秒前
ahuang2222完成签到,获得积分10
24秒前
苏州小燕子完成签到,获得积分20
27秒前
27秒前
32秒前
33秒前
Trailblazer完成签到,获得积分10
33秒前
Hello应助潘宇霜采纳,获得10
38秒前
MiuMiu发布了新的文献求助10
38秒前
CynthiaaaCat发布了新的文献求助10
38秒前
学术混子发布了新的文献求助10
39秒前
39秒前
39秒前
tony完成签到,获得积分10
39秒前
wjx完成签到,获得积分10
40秒前
42秒前
wjx发布了新的文献求助10
43秒前
43秒前
小白发布了新的文献求助10
45秒前
xiuwenli发布了新的文献求助10
47秒前
49秒前
科研狗发布了新的文献求助20
54秒前
打打应助深情的牛排采纳,获得10
57秒前
高分求助中
Sustainable Land Management: Strategies to Cope with the Marginalisation of Agriculture 1000
Corrosion and Oxygen Control 600
Yaws' Handbook of Antoine coefficients for vapor pressure 500
Python Programming for Linguistics and Digital Humanities: Applications for Text-Focused Fields 500
Division and square root. Digit-recurrence algorithms and implementations 400
行動データの計算論モデリング 強化学習モデルを例として 400
Johann Gottlieb Fichte: Die späten wissenschaftlichen Vorlesungen / IV,1: ›Transzendentale Logik I (1812)‹ 400
热门求助领域 (近24小时)
化学 材料科学 医学 生物 有机化学 工程类 生物化学 纳米技术 物理 内科学 计算机科学 化学工程 复合材料 遗传学 基因 物理化学 催化作用 电极 光电子学 量子力学
热门帖子
关注 科研通微信公众号,转发送积分 2552998
求助须知:如何正确求助?哪些是违规求助? 2178405
关于积分的说明 5614205
捐赠科研通 1899419
什么是DOI,文献DOI怎么找? 948387
版权声明 565554
科研通“疑难数据库(出版商)”最低求助积分说明 504370