Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(7): 1589-1616, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38552609

ABSTRACT

The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.


Subject(s)
Neoplasms , Humans , Carcinogenesis , Microbiota , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Obesity/complications , Quality of Life
2.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649199

ABSTRACT

Interleukin-1ß (IL-1ß)-mediated inflammation suppresses antitumor immunity, leading to the generation of a tumor-permissive environment, tumor growth, and progression. Here, we demonstrate that nucleotide-binding domain, leucine-rich containing family, pyrin domain-containing-3 (NLRP3) inflammasome activation in melanoma is linked to IL-1ß production, inflammation, and immunosuppression. Analysis of cancer genome datasets (TCGA and GTEx) revealed greater NLRP3 and IL-1ß expression in cutaneous melanoma samples (n = 469) compared to normal skin (n = 324), with a highly significant correlation between NLRP3 and IL-1ß (P < 0.0001). We show the formation of the NLRP3 inflammasome in biopsies of metastatic melanoma using fluorescent resonance energy transfer analysis for NLRP3 and apoptosis-associated speck-like protein containing a CARD. In vivo, tumor-associated NLRP3/IL-1 signaling induced expansion of myeloid-derived suppressor cells (MDSCs), leading to reduced natural killer and CD8+ T cell activity concomitant with an increased presence of regulatory T (Treg) cells in the primary tumors. Either genetic or pharmacological inhibition of tumor-derived NLRP3 by dapansutrile (OLT1177) was sufficient to reduce MDSCs expansion and to enhance antitumor immunity, resulting in reduced tumor growth. Additionally, we observed that the combination of NLRP3 inhibition and anti-PD-1 treatment significantly increased the antitumor efficacy of the monotherapy by limiting MDSC-mediated T cell suppression and tumor progression. These data show that NLRP3 activation in melanoma cells is a protumor mechanism, which induces MDSCs expansion and immune evasion. We conclude that inhibition of NLRP3 can augment the efficacy of anti-PD-1 therapy.


Subject(s)
Melanoma, Experimental/immunology , Myeloid-Derived Suppressor Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neoplasm Proteins/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neoplasm Proteins/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology
3.
4.
Mol Biol Evol ; 37(2): 320-326, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31642480

ABSTRACT

Cancer progression is an evolutionary process. During this process, evolving cancer cell populations encounter restrictive ecological niches within the body, such as the primary tumor, circulatory system, and diverse metastatic sites. Efforts to prevent or delay cancer evolution-and progression-require a deep understanding of the underlying molecular evolutionary processes. Herein we discuss a suite of concepts and tools from evolutionary and ecological theory that can inform cancer biology in new and meaningful ways. We also highlight current challenges to applying these concepts, and propose ways in which incorporating these concepts could identify new therapeutic modes and vulnerabilities in cancer.


Subject(s)
Genomics/methods , Neoplasms/genetics , Disease Progression , Evolution, Molecular , Genetic Fitness , Humans , Phylogeny , Stem Cell Niche
5.
Blood ; 134(4): 389-394, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31101624

ABSTRACT

We have previously demonstrated that oxidative phosphorylation is required for the survival of human leukemia stem cells (LSCs) from patients with acute myeloid leukemia (AML). More recently, we demonstrated that LSCs in patients with de novo AML rely on amino acid metabolism to drive oxidative phosphorylation. Notably, although overall levels of amino acids contribute to LSC energy metabolism, our current findings suggest that cysteine may be of particular importance for LSC survival. We demonstrate that exogenous cysteine is metabolized exclusively to glutathione. Upon cysteine depletion, glutathione synthesis is impaired, leading to reduced glutathionylation of succinate dehydrogenase A (SDHA), a key component of electron transport chain complex (ETC) II. Loss of SDHA glutathionylation impairs ETC II activity, thereby inhibiting oxidative phosphorylation, reducing production of ATP, and leading to LSC death. Given the role of cysteine in driving LSC energy production, we tested cysteine depletion as a potential therapeutic strategy. Using a novel cysteine-degrading enzyme, we demonstrate selective eradication of LSCs, with no detectable effect on normal hematopoietic stem/progenitor cells. Together, these findings indicate that LSCs are aberrantly reliant on cysteine to sustain energy metabolism, and that targeting this axis may represent a useful therapeutic strategy.


Subject(s)
Cysteine/metabolism , Electron Transport Complex II/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Adenosine Triphosphate/metabolism , Biomarkers , Energy Metabolism , Glutathione/metabolism , Humans , Oxidation-Reduction , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism , Succinate Dehydrogenase/metabolism
6.
Blood ; 134(9): 727-740, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31311815

ABSTRACT

Aging and chronic inflammation are independent risk factors for the development of atherothrombosis and cardiovascular disease. We hypothesized that aging-associated inflammation promotes the development of platelet hyperreactivity and increases thrombotic risk during aging. Functional platelet studies in aged-frail adults and old mice demonstrated that their platelets are hyperreactive and form larger thrombi. We identified tumor necrosis factor α (TNF-α) as the key aging-associated proinflammatory cytokine responsible for platelet hyperreactivity. We further showed that platelet hyperreactivity is neutralized by abrogating signaling through TNF-α receptors in vivo in a mouse model of aging. Analysis of the bone marrow compartments showed significant platelet-biased hematopoiesis in old mice reflected by increased megakaryocyte-committed progenitor cells, megakaryocyte ploidy status, and thrombocytosis. Single-cell RNA-sequencing analysis of native mouse megakaryocytes showed significant reprogramming of inflammatory, metabolic, and mitochondrial gene pathways in old mice that appeared to play a significant role in determining platelet hyperreactivity. Platelets from old mice (where TNF-α was endogenously increased) and from young mice exposed to exogenous TNF-α exhibited significant mitochondrial changes characterized by elevated mitochondrial mass and increased oxygen consumption during activation. These mitochondrial changes were mitigated upon TNF-α blockade. Similar increases in platelet mitochondrial mass were seen in platelets from patients with myeloproliferative neoplasms, where TNF-α levels are also increased. Furthermore, metabolomics studies of platelets from young and old mice demonstrated age-dependent metabolic profiles that may differentially poise platelets for activation. Altogether, we present previously unrecognized evidence that TNF-α critically regulates megakaryocytes resident in the bone marrow niche and aging-associated platelet hyperreactivity and thrombosis.


Subject(s)
Aging , Blood Platelets/immunology , Inflammation/immunology , Mitochondria/immunology , Thrombosis/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Blood Platelets/pathology , Inflammation/pathology , Megakaryocytes/immunology , Megakaryocytes/pathology , Mice , Mice, Inbred C57BL , Mitochondria/pathology , Platelet Activation , Thrombosis/pathology
7.
Br J Cancer ; 122(7): 943-952, 2020 03.
Article in English | MEDLINE | ID: mdl-32042067

ABSTRACT

Why do we get cancer mostly when we are old? According to current paradigms, the answer is simple: mutations accumulate in our tissues throughout life, and some of these mutations contribute to cancers. Although mutations are necessary for cancer development, a number of studies shed light on roles for ageing and exposure-dependent changes in tissue landscapes that determine the impact of oncogenic mutations on cellular fitness, placing carcinogenesis into an evolutionary framework. Natural selection has invested in somatic maintenance to maximise reproductive success. Tissue maintenance not only ensures functional robustness but also prevents the occurrence of cancer through periods of likely reproduction by limiting selection for oncogenic events in our cells. Indeed, studies in organisms ranging from flies to humans are revealing conserved mechanisms to eliminate damaged or oncogenically initiated cells from tissues. Reports of the existence of striking numbers of oncogenically initiated clones in normal tissues and of how this clonal architecture changes with age or external exposure to noxious substances provide critical insight into the early stages of cancer development. A major challenge for cancer biology will be the integration of these studies with epidemiology data into an evolutionary theory of carcinogenesis, which could have a large impact on addressing cancer risk and treatment.


Subject(s)
Aging/pathology , Tumor Microenvironment/genetics , Aged , Aged, 80 and over , Biological Evolution , Female , Humans , Male , Middle Aged , Mutation , Neoplasms
8.
Biochim Biophys Acta Rev Cancer ; 1867(2): 84-94, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28167050

ABSTRACT

By the time the process of oncogenesis has produced an advanced cancer, tumor cells have undergone extensive evolution. The cellular phenotypes resulting from this evolution have been well studied, and include accelerated growth rates, apoptosis resistance, immortality, invasiveness, and immune evasion. Yet with all of our current knowledge of tumor biology, the details of early oncogenesis have been difficult to observe and understand. Where different oncogenic mutations may work together to enhance the survival of a tumor cell, in isolation they are often pro-apoptotic, pro-differentiative or pro-senescent, and therefore often, somewhat paradoxically, disadvantageous to a cell. It is also becoming clear that somatic mutations, including those in known oncogenic drivers, are common in tissues starting at a young age. These observations raise the question: how do we largely avoid cancer for most of our lives? Here we propose that evolutionary forces can help explain this paradox. As humans and other organisms age or experience external insults such as radiation or smoking, the structure and function of tissues progressively degrade, resulting in altered stem cell niche microenvironments. As tissue integrity declines, it becomes less capable of supporting and maintaining resident stem cells. These stem cells then find themselves in a microenvironment to which they are poorly adapted, providing a competitive advantage to those cells that can restore their functionality and fitness through mutations or epigenetic changes. The resulting oncogenic clonal expansions then increase the odds of further cancer progression. Understanding how the causes of cancer, such as aging or smoking, affect tissue microenvironments to control the impact of mutations on somatic cell fitness can help reconcile the discrepancy between marked mutation accumulation starting early in life and the somatic evolution that leads to cancer at advanced ages or following carcinogenic insults. This article is part of a Special Issue entitled: Evolutionary principles - heterogeneity in cancer?, edited by Dr. Robert A. Gatenby.


Subject(s)
Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Evolution, Molecular , Genetic Fitness , Mutation , Neoplasms/genetics , Adaptation, Physiological , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Heredity , Humans , Models, Genetic , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Pedigree , Phenotype , Signal Transduction/genetics , Time Factors
9.
Nature ; 568(7752): 318-319, 2019 04.
Article in English | MEDLINE | ID: mdl-30980033
10.
BMC Evol Biol ; 19(1): 172, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31443631

ABSTRACT

BACKGROUND: The evolution of multi-cellular animals has produced a conspicuous trend toward increased body size. This trend has introduced at least two novel problems: an expected elevated risk of somatic disorders, such as cancer, and declining evolvability due to generally reduced population size, lower reproduction rate and extended generation time. Low population size is widely recognized to explain the high mutation rates in animals by limiting the presumed universally negative selection acting on mutation rates. RESULTS: Here, we present evidence from stochastic modeling that the direction and strength of selection acting on mutation rates is highly dependent on the evolution of somatic maintenance, and thus longevity, which modulates the cost of somatic mutations. CONCLUSIONS: We argue that the impact of the evolution of longevity on mutation rates may have been critical in facilitating animal evolution.


Subject(s)
Biological Evolution , Body Size , Models, Genetic , Mutation Rate , Animals , Computer Simulation , Longevity , Monte Carlo Method , Mutation , Neoplasms/genetics , Phenotype , Population Density , Selection, Genetic
12.
Proc Natl Acad Sci U S A ; 113(4): 1050-5, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26755588

ABSTRACT

Young children have higher rates of leukemia than young adults. This fact represents a fundamental conundrum, because hematopoietic cells in young children should have fewer mutations (including oncogenic ones) than such cells in adults. Here, we present the results of stochastic modeling of hematopoietic stem cell (HSC) clonal dynamics, which demonstrated that early HSC pools were permissive to clonal evolution driven by drift. We show that drift-driven clonal expansions cooperate with faster HSC cycling in young children to produce conditions that are permissive for accumulation of multiple driver mutations in a single cell. Later in life, clonal evolution was suppressed by stabilizing selection in the larger young adult pools, and it was driven by positive selection at advanced ages in the presence of microenvironmental decline. Overall, our results indicate that leukemogenesis is driven by distinct evolutionary forces in children and adults.


Subject(s)
Clonal Evolution , Leukemia/etiology , Age Factors , Child , Hematopoietic Stem Cells/physiology , Humans , Mutation , Stochastic Processes
13.
Proc Natl Acad Sci U S A ; 113(43): E6669-E6678, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27791036

ABSTRACT

Activating mutations in FMS-like tyrosine kinase 3 (FLT3) are common in acute myeloid leukemia (AML) and drive leukemic cell growth and survival. Although FLT3 inhibitors have shown considerable promise for the treatment of AML, they ultimately fail to achieve long-term remissions as monotherapy. To identify genetic targets that can sensitize AML cells to killing by FLT3 inhibitors, we performed a genome-wide RNA interference (RNAi)-based screen that identified ATM (ataxia telangiectasia mutated) as being synthetic lethal with FLT3 inhibitor therapy. We found that inactivating ATM or its downstream effector glucose 6-phosphate dehydrogenase (G6PD) sensitizes AML cells to FLT3 inhibitor induced apoptosis. Examination of the cellular metabolome showed that FLT3 inhibition by itself causes profound alterations in central carbon metabolism, resulting in impaired production of the antioxidant factor glutathione, which was further impaired by ATM or G6PD inactivation. Moreover, FLT3 inhibition elicited severe mitochondrial oxidative stress that is causative in apoptosis and is exacerbated by ATM/G6PD inhibition. The use of an agent that intensifies mitochondrial oxidative stress in combination with a FLT3 inhibitor augmented elimination of AML cells in vitro and in vivo, revealing a therapeutic strategy for the improved treatment of FLT3 mutated AML.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Leukemic , Glucosephosphate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Benzothiazoles/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Therapy, Combination , Female , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Glucosephosphate Dehydrogenase/metabolism , Humans , Hydrazines/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice, Inbred NOD , Middle Aged , Oxidation-Reduction , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/metabolism
14.
Nucleic Acids Res ; 44(5): 2075-84, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26883632

ABSTRACT

Mutations acquired during development and aging lead to inter- and intra-tissue genetic variations. Evidence linking such mutations to complex traits and diseases is rising. We detected somatic mutations in protein-coding regions in 140 benign tissue samples representing nine tissue-types (bladder, breast, liver, lung, prostate, stomach, thyroid, head and neck) and paired blood from 70 donors. A total of 80% of the samples had 2-39 mutations detectable at tissue-level resolution. Factors such as age and smoking were associated with increased burden of detectable mutations, and tissues carried signatures of distinct mutagenic processes such as oxidative DNA damage and transcription-coupled repair. Using mutational signatures, we predicted that majority of the mutations in blood originated in hematopoietic stem and early progenitor cells. Missense to silent mutations ratio and the persistence of potentially damaging mutations in expressed genes carried signatures of relaxed purifying selection. Our findings have relevance for etiology, diagnosis and treatment of diseases including cancer.


Subject(s)
Aging/genetics , Exome , Neoplasms/genetics , Open Reading Frames , Selection, Genetic , Tumor Microenvironment/genetics , Female , Genome, Human , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Models, Genetic , Mutation Rate , Mutation, Missense , Neoplasms/pathology , Organ Specificity
15.
Proc Natl Acad Sci U S A ; 112(29): 8914-21, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26195756

ABSTRACT

Our understanding of cancer has greatly advanced since Nordling [Nordling CO (1953) Br J Cancer 7(1):68-72] and Armitage and Doll [Armitage P, Doll R (1954) Br J Cancer 8(1):1-12] put forth the multistage model of carcinogenesis. However, a number of observations remain poorly understood from the standpoint of this paradigm in its contemporary state. These observations include the similar age-dependent exponential rise in incidence of cancers originating from stem/progenitor pools differing drastically in size, age-dependent cell division profiles, and compartmentalization. This common incidence pattern is characteristic of cancers requiring different numbers of oncogenic mutations, and it scales to very divergent life spans of mammalian species. Also, bigger mammals with larger underlying stem cell pools are not proportionally more prone to cancer, an observation known as Peto's paradox. Here, we present a number of factors beyond the occurrence of oncogenic mutations that are unaccounted for in the current model of cancer development but should have significant impacts on cancer incidence. Furthermore, we propose a revision of the current understanding for how oncogenic and other functional somatic mutations affect cellular fitness. We present evidence, substantiated by evolutionary theory, demonstrating that fitness is a dynamic environment-dependent property of a phenotype and that oncogenic mutations should have vastly different fitness effects on somatic cells dependent on the tissue microenvironment in an age-dependent manner. Combined, this evidence provides a firm basis for understanding the age-dependent incidence of cancers as driven by age-altered systemic processes regulated above the cell level.


Subject(s)
Biological Evolution , Models, Biological , Mutation/genetics , Neoplasms/genetics , Animals , Genetic Drift , Genetic Fitness , Humans , Nonlinear Dynamics , Selection, Genetic , Stem Cells/metabolism , Time Factors
16.
Blood ; 136(2): 153-154, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32645167
17.
Haematologica ; 102(12): 1985-1994, 2017 12.
Article in English | MEDLINE | ID: mdl-28883079

ABSTRACT

While dietary folate deficiency is associated with increased risk for birth defects and other diseases, evidence suggests that supplementation with folic acid can contribute to predisposition to some diseases, including immune dysfunction and cancer. Herein, we show that diets supplemented with folic acid both below and above the recommended levels led to significantly altered metabolism in multiple tissues in mice. Surprisingly, both low and excessive dietary folate induced similar metabolic changes, which were particularly evident for nucleotide biosynthetic pathways in B-progenitor cells. Diet-induced metabolic changes in these cells partially phenocopied those observed in mice treated with anti-folate drugs, suggesting that both deficiency and excessive levels of dietary folic acid compromise folate-dependent biosynthetic pathways. Both folate deficiency and excessive dietary folate levels compromise hematopoiesis, resulting in defective cell cycle progression, persistent DNA damage, and impaired production of lymphocytes. These defects reduce the reconstitution potential in transplantation settings and increase radiation-induced mortality. We conclude that excessive folic acid supplementation can metabolically mimic dietary folate insufficiency, leading to similar functional impairment of hematopoiesis.


Subject(s)
Dietary Supplements/adverse effects , Folic Acid Deficiency/metabolism , Folic Acid/pharmacology , Hematopoiesis/drug effects , Animals , Folic Acid/metabolism , Folic Acid/therapeutic use , Metabolism/drug effects , Mice , Nucleotides/biosynthesis , Precursor Cells, B-Lymphoid/drug effects , Precursor Cells, B-Lymphoid/metabolism
18.
PLoS Genet ; 10(3): e1004239, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24603726

ABSTRACT

Cancer is an evolutionary process in which cells acquire new transformative, proliferative and metastatic capabilities. A full understanding of cancer requires learning the dynamics of the cancer evolutionary process. We present here a large-scale analysis of the dynamics of this evolutionary process within tumors, with a focus on breast cancer. We show that the cancer evolutionary process differs greatly from organismal (germline) evolution. Organismal evolution is dominated by purifying selection (that removes mutations that are harmful to fitness). In contrast, in the cancer evolutionary process the dominance of purifying selection is much reduced, allowing for a much easier detection of the signals of positive selection (adaptation). We further show that, as a group, genes that are globally expressed across human tissues show a very strong signal of positive selection within tumors. Indeed, known cancer genes are enriched for global expression patterns. Yet, positive selection is prevalent even on globally expressed genes that have not yet been associated with cancer, suggesting that globally expressed genes are enriched for yet undiscovered cancer related functions. We find that the increased positive selection on globally expressed genes within tumors is not due to their expression in the tissue relevant to the cancer. Rather, such increased adaptation is likely due to globally expressed genes being enriched in important housekeeping and essential functions. Thus, our results suggest that tumor adaptation is most often mediated through somatic changes to those genes that are important for the most basic cellular functions. Together, our analysis reveals the uniqueness of the cancer evolutionary process and the particular importance of globally expressed genes in driving cancer initiation and progression.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Evolution, Molecular , Selection, Genetic , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation
19.
J Biol Chem ; 290(21): 13479-89, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25847239

ABSTRACT

Protein arginine methyl transferase 1 (PRMT1) was shown to be up-regulated in cancers and important for cancer cell proliferation. However, the role of PRMT1 in lung cancer progression and metastasis remains incompletely understood. In the present study, we show that PRMT1 is an important regulator of epithelial-mesenchymal transition (EMT), cancer cell migration, and invasion, which are essential processes during cancer progression, and metastasis. Additionally, we have identified Twist1, a basic helix-loop-helix transcription factor and a well-known E-cadherin repressor, as a novel PRMT1 substrate. Taken together, we show that PRMT1 is a novel regulator of EMT and arginine 34 (Arg-34) methylation of Twist1 as a unique "methyl arginine mark" for active E-cadherin repression. Therefore, targeting PRMT1-mediated Twist1 methylation might represent a novel strategy for developing new anti-invasive/anti-metastatic drugs. Moreover, methylated Twist1 (Arg-34), as such, could also emerge as a potential important biomarker for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/secondary , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Arginine/genetics , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation , DNA Methylation , Fluorescent Antibody Technique, Indirect , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Molecular Sequence Data , Neoplasm Invasiveness , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Tumor Cells, Cultured , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Wound Healing , Xenograft Model Antitumor Assays
20.
Stem Cells ; 33(4): 1345-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25546133

ABSTRACT

Ionizing radiation (IR) is associated with reduced hematopoietic function and increased risk of hematopoietic malignancies, although the mechanisms behind these relationships remain poorly understood. Both effects of IR have been commonly attributed to the direct induction of DNA mutations, but evidence supporting these hypotheses is largely lacking. Here we demonstrate that IR causes long-term, somatically heritable, cell-intrinsic reductions in hematopoietic stem cell (HSC) and multipotent hematopoietic progenitor cell (mHPC) self-renewal that are mediated by C/EBPα and reversed by Notch. mHPC from previously irradiated (>9 weeks prior), homeostatically restored mice exhibit gene expression profiles consistent with their precocious differentiation phenotype, including decreased expression of HSC-specific genes and increased expression of myeloid program genes (including C/EBPα). These gene expression changes are reversed by ligand-mediated activation of Notch. Loss of C/EBPα expression is selected for within previously irradiated HSC and mHPC pools and is associated with reversal of IR-dependent precocious differentiation and restoration of self-renewal. Remarkably, restoration of mHPC self-renewal by ligand-mediated activation of Notch prevents selection for C/EBPα loss of function in previously irradiated mHPC pools. We propose that environmental insults prompt HSC to initiate a program limiting their self-renewal, leading to loss of the damaged HSC from the pool while allowing this HSC to temporarily contribute to differentiated cell pools. This "programmed mediocrity" is advantageous for the sporadic genotoxic insults animals have evolved to deal with but becomes tumor promoting when the entire HSC compartment is damaged, such as during total body irradiation, by increasing selective pressure for adaptive oncogenic mutations.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/radiation effects , Hematopoietic Stem Cells/radiation effects , Multipotent Stem Cells/radiation effects , Radiation, Ionizing , Receptors, Notch/radiation effects , Animals , CCAAT-Enhancer-Binding Protein-alpha/physiology , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cell Proliferation/physiology , Cell Proliferation/radiation effects , Cells, Cultured , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multipotent Stem Cells/physiology , Receptors, Notch/physiology
SELECTION OF CITATIONS
SEARCH DETAIL