Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
CA Cancer J Clin ; 67(4): 326-344, 2017 07 08.
Article in English | MEDLINE | ID: mdl-28481406

ABSTRACT

Answer questions and earn CME/CNE The human body harbors enormous numbers of microbiota that influence cancer susceptibility, in part through their prodigious metabolic capacity and their profound influence on immune cell function. Microbial pathogens drive tumorigenesis in 15% to 20% of cancer cases. Even larger numbers of malignancies are associated with an altered composition of commensal microbiota (dysbiosis) based on microbiome studies using metagenomic sequencing. Although association studies cannot distinguish whether changes in microbiota are causes or effects of cancer, a causative role is supported by rigorously controlled preclinical studies using gnotobiotic mouse models colonized with one or more specific bacteria. These studies demonstrate that microbiota can alter cancer susceptibility and progression by diverse mechanisms, such as modulating inflammation, inducing DNA damage, and producing metabolites involved in oncogenesis or tumor suppression. Evidence is emerging that microbiota can be manipulated for improving cancer treatment. By incorporating probiotics as adjuvants for checkpoint immunotherapy or by designing small molecules that target microbial enzymes, microbiota can be harnessed to improve cancer care. CA Cancer J Clin 2017;67:326-344. © 2017 American Cancer Society.


Subject(s)
Microbiota , Neoplasms/microbiology , Neoplasms/therapy , Animals , Carcinogenesis , Disease Models, Animal , Disease Progression , Disease Susceptibility , Dysbiosis , Humans , Metagenomics , Precision Medicine
2.
Proc Natl Acad Sci U S A ; 117(13): 7374-7381, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32170007

ABSTRACT

Irinotecan treats a range of solid tumors, but its effectiveness is severely limited by gastrointestinal (GI) tract toxicity caused by gut bacterial ß-glucuronidase (GUS) enzymes. Targeted bacterial GUS inhibitors have been shown to partially alleviate irinotecan-induced GI tract damage and resultant diarrhea in mice. Here, we unravel the mechanistic basis for GI protection by gut microbial GUS inhibitors using in vivo models. We use in vitro, in fimo, and in vivo models to determine whether GUS inhibition alters the anticancer efficacy of irinotecan. We demonstrate that a single dose of irinotecan increases GI bacterial GUS activity in 1 d and reduces intestinal epithelial cell proliferation in 5 d, both blocked by a single dose of a GUS inhibitor. In a tumor xenograft model, GUS inhibition prevents intestinal toxicity and maintains the antitumor efficacy of irinotecan. Remarkably, GUS inhibitor also effectively blocks the striking irinotecan-induced bloom of Enterobacteriaceae in immune-deficient mice. In a genetically engineered mouse model of cancer, GUS inhibition alleviates gut damage, improves survival, and does not alter gut microbial composition; however, by allowing dose intensification, it dramatically improves irinotecan's effectiveness, reducing tumors to a fraction of that achieved by irinotecan alone, while simultaneously promoting epithelial regeneration. These results indicate that targeted gut microbial enzyme inhibitors can improve cancer chemotherapeutic outcomes by protecting the gut epithelium from microbial dysbiosis and proliferative crypt damage.


Subject(s)
Gastrointestinal Microbiome/drug effects , Glucuronidase/antagonists & inhibitors , Glucuronidase/drug effects , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Bacteria/drug effects , Disease Models, Animal , Dysbiosis/drug therapy , Enzyme Inhibitors/pharmacology , Female , Glucuronidase/metabolism , Humans , Irinotecan/pharmacology , Mice , Mice, Nude , Neoplasms/drug therapy
3.
Mol Cell ; 48(4): 612-26, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23063526

ABSTRACT

Widespread changes in gene expression drive tumorigenesis, yet our knowledge of how aberrant epigenomic and transcriptome profiles arise in cancer cells is poorly understood. Here, we demonstrate that metabolic transformation plays an important role. Butyrate is the primary energy source of normal colonocytes and is metabolized to acetyl-CoA, which was shown to be important not only for energetics but also for HAT activity. Due to the Warburg effect, cancerous colonocytes rely on glucose as their primary energy source, so butyrate accumulated and functioned as an HDAC inhibitor. Although both mechanisms increased histone acetylation, different target genes were upregulated. Consequently, butyrate stimulated the proliferation of normal colonocytes and cancerous colonocytes when the Warburg effect was prevented from occurring, whereas it inhibited the proliferation of cancerous colonocytes undergoing the Warburg effect. These findings link a common metabolite to epigenetic mechanisms that are differentially utilized by normal and cancerous cells because of their inherent metabolic differences.


Subject(s)
Butyrates/metabolism , Histones/metabolism , Models, Biological , Acetylation , Cell Proliferation , HCT116 Cells , HT29 Cells , Humans , Tumor Cells, Cultured
4.
J Mol Cell Cardiol ; 105: 99-109, 2017 04.
Article in English | MEDLINE | ID: mdl-28232072

ABSTRACT

RATIONALE: The contractile dysfunction that underlies heart failure involves perturbations in multiple biological processes ranging from metabolism to electrophysiology. Yet the epigenetic mechanisms that are altered in this disease state have not been elucidated. SWI/SNF chromatin-remodeling complexes are plausible candidates based on mouse knockout studies demonstrating a combined requirement for the BRG1 and BRM catalytic subunits in adult cardiomyocytes. Brg1/Brm double mutants exhibit metabolic and mitochondrial defects and are not viable although their cause of death has not been ascertained. OBJECTIVE: To determine the cause of death of Brg1/Brm double-mutant mice, to test the hypothesis that BRG1 and BRM are required for cardiac contractility, and to identify relevant downstream target genes. METHODS AND RESULTS: A tamoxifen-inducible gene-targeting strategy utilizing αMHC-Cre-ERT was implemented to delete both SWI/SNF catalytic subunits in adult cardiomyocytes. Brg1/Brm double-mutant mice were monitored by echocardiography and electrocardiography, and they underwent rapidly progressive ventricular dysfunction including conduction defects and arrhythmias that culminated in heart failure and death within 3weeks. Mechanistically, BRG1/BRM repressed c-Myc expression, and enforced expression of a DOX-inducible c-MYC trangene in mouse cardiomyocytes phenocopied the ventricular conduction defects observed in Brg1/Brm double mutants. BRG1/BRM and c-MYC had opposite effects on the expression of cardiac conduction genes, and the directionality was consistent with their respective loss- and gain-of-function phenotypes. To support the clinical relevance of this mechanism, BRG1/BRM occupancy was diminished at the same target genes in human heart failure cases compared to controls, and this correlated with increased c-MYC expression and decreased CX43 and SCN5A expression. CONCLUSION: BRG1/BRM and c-MYC have an antagonistic relationship regulating the expression of cardiac conduction genes that maintain contractility, which is reminiscent of their antagonistic roles as a tumor suppressor and oncogene in cancer.


Subject(s)
DNA Helicases/metabolism , Heart Conduction System , Myocardial Contraction , Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Animals , DNA Helicases/genetics , Electrocardiography , Gene Expression Profiling , Gene Expression Regulation , Heart Failure/diagnosis , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Mice , Mice, Transgenic , Mutation , Myocardial Contraction/genetics , Nuclear Proteins/genetics , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics
5.
EMBO Rep ; 16(8): 1037-50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26136374

ABSTRACT

Although the two catalytic subunits of the SWI/SNF chromatin-remodeling complex--Brahma (Brm) and Brg1--are almost invariably co-expressed, their mutually exclusive incorporation into distinct SWI/SNF complexes predicts that Brg1- and Brm-based SWI/SNF complexes execute specific functions. Here, we show that Brg1 and Brm have distinct functions at discrete stages of muscle differentiation. While Brg1 is required for the activation of muscle gene transcription at early stages of differentiation, Brm is required for Ccnd1 repression and cell cycle arrest prior to the activation of muscle genes. Ccnd1 knockdown rescues the ability to exit the cell cycle in Brm-deficient myoblasts, but does not recover terminal differentiation, revealing a previously unrecognized role of Brm in the activation of late muscle gene expression independent from the control of cell cycle. Consistently, Brm null mice displayed impaired muscle regeneration after injury, with aberrant proliferation of satellite cells and delayed formation of new myofibers. These data reveal stage-specific roles of Brm during skeletal myogenesis, via formation of repressive and activatory SWI/SNF complexes.


Subject(s)
Cell Cycle Checkpoints/genetics , DNA Helicases/metabolism , Gene Expression , Muscle Development/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cyclin D1/deficiency , Cyclin D1/genetics , DNA Helicases/genetics , Gene Knockdown Techniques , Mice , Muscle Cells , Nuclear Proteins/genetics , Transcription Factors/genetics
6.
Carcinogenesis ; 35(2): 249-55, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24302613

ABSTRACT

Gene-environment interactions underlie cancer susceptibility and progression. Yet, we still have limited knowledge of which environmental factors are important and how they function during tumorigenesis. In this respect, the microbial communities that inhabit our gastrointestinal tract and other body sites have been unappreciated until recently. However, our microbiota are environmental factors that we are exposed to continuously, and human microbiome studies have revealed significant differences in the relative abundance of certain microbes in cancer cases compared with controls. To characterize the function of microbiota in carcinogenesis, mouse models of cancer have been treated with antibiotics. They have also been maintained in a germfree state or have been colonized with specific bacteria in specialized (gnotobiotic) facilities. These studies demonstrate that microbiota can increase or decrease cancer susceptibility and progression by diverse mechanisms such as by modulating inflammation, influencing the genomic stability of host cells and producing metabolites that function as histone deacetylase inhibitors to epigenetically regulate host gene expression. One might consider microbiota as tractable environmental factors because they are highly quantifiable and relatively stable within an individual compared with our exposures to external agents. At the same time, however, diet can modulate the composition of microbial communities within our gut, and this supports the idea that probiotics and prebiotics can be effective chemoprevention strategies. The trajectory of where the current work is headed suggests that microbiota will continue to provide insight into the basic mechanisms of carcinogenesis and that microbiota will also become targets for therapeutic intervention.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Gastrointestinal Tract/microbiology , Microbiota/drug effects , Neoplasms/microbiology , Animals , Gastrointestinal Tract/drug effects , Humans , Mice , Neoplasms/drug therapy
7.
J Cell Physiol ; 229(1): 44-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23757284

ABSTRACT

Widespread changes in gene expression underlie B cell development and activation, yet our knowledge of which chromatin-remodeling factors are essential is limited. Here, we demonstrate that the BRG1 catalytic subunit of SWI/SNF complexes was dispensable for murine B cell development but played an important, albeit selective, role during activation. Although BRG1 was dispensable for CD69 induction and differentiation into plasma cells based on the ability of mutant B cells to undergo hypertrophy and secrete IgM antibodies, it was required for robust cell proliferation in response to activation. Accordingly, BRG1 was required for only ∼100 genes to be expressed at normal levels in naïve B cells but >1,000 genes during their activation. BRG1 upregulated fivefold more genes than it downregulated, and the toll-like receptor pathway and JAK/STAT cytokine-signaling pathways were particularly dependent on BRG1. The importance of BRG1 in B cell activation was underscored by the occurrence of opportunistic Pasteurella infections in conditionally mutant mice. B cell activation has long served as a model of inducible gene expression, and the results presented here identify BRG1 as a chromatin-remodeling factor that upregulates the transcriptome of B cells during their activation to promote rapid cell proliferation and to mount an effective immune response.


Subject(s)
B-Lymphocytes/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Helicases , Lymphocyte Activation/genetics , Nuclear Proteins , Transcription Factors , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Cell Differentiation/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
8.
Circ Res ; 111(5): e111-22, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22740088

ABSTRACT

RATIONALE: Mating type switching/sucrose non-fermenting (SWI/SNF) chromatin-remodeling complexes utilize either BRG1 or BRM as a catalytic subunit to alter nucleosome position and regulate gene expression. BRG1 is required for vascular endothelial cell (VEC) development and embryonic survival, whereas BRM is dispensable. OBJECTIVE: To circumvent embryonic lethality and study Brg1 function in adult tissues, we used conditional gene targeting. To evaluate possible Brg1-Brm redundancy, we analyzed Brg1 mutant mice on wild-type and Brm-deficient backgrounds. METHODS AND RESULTS: The inducible Mx1-Cre driver was used to mutate Brg1 in adult mice. These conditional-null mutants exhibited a tissue-specific phenotype and unanticipated functional compensation between Brg1 and Brm. Brg1 single mutants were healthy and had a normal lifespan, whereas Brg1/Brm double mutants exhibited cardiovascular defects and died within 1 month. BRG1 and BRM were required for the viability of VECs but not other cell types where both genes were also knocked out. The VEC phenotype was most evident in the heart, particularly in the microvasculature of the outer myocardium, and was recapitulated in primary cells ex vivo. VEC death resulted in vascular leakage, cardiac hemorrhage, secondary death of cardiomyocytes due to ischemia, and ventricular dissections. CONCLUSIONS: BRG1-catalyzed SWI/SNF complexes are particularly important in cardiovascular tissues. However, in contrast to embryonic development, in which Brm does not compensate, Brg1 is required in adult VECs only when Brm is also mutated. These results demonstrate for the first time that Brm functionally compensates for Brg1 in vivo and that there are significant changes in the relative importance of BRG1- and BRM-catalyzed SWI/SNF complexes during the development of an essential cell lineage.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , Endothelial Cells/metabolism , Heart Defects, Congenital/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Age Factors , Animals , Catalysis , Cell Death/physiology , Cell Lineage/physiology , Cell Survival/physiology , Chromosomal Proteins, Non-Histone/genetics , Coronary Vessels/embryology , Coronary Vessels/metabolism , Coronary Vessels/pathology , DNA Helicases/genetics , Echocardiography , Endothelial Cells/pathology , Heart/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Homeostasis/physiology , Mice , Mice, Transgenic , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Nuclear Proteins/genetics , Pleural Effusion/genetics , Pleural Effusion/metabolism , Pleural Effusion/pathology , Transcription Factors/genetics
9.
Mamm Genome ; 24(1-2): 21-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23076393

ABSTRACT

Mammalian SWI/SNF complexes utilize either BRG1 or BRM as alternative catalytic subunits with DNA-dependent ATPase activity to remodel chromatin. Although the two proteins are 75 % identical, broadly expressed, and have similar biochemical activities in vitro, BRG1 is essential for mouse embryonic development, while BRM is dispensable. To investigate whether BRG1 and BRM have overlapping functions during mouse embryogenesis, we performed double-heterozygous intercrosses using constitutive null mutations previously created by gene targeting. The progeny of these crosses had a distribution of genotypes that was significantly skewed relative to their combined gene dosage. This was most pronounced at the top and bottom of the gene dosage hierarchy, with a 1.5-fold overrepresentation of Brg1 (+/+) ;Brm (+/+) mice and a corresponding 1.6-fold underrepresentation of Brg1 (+/-) ;Brm (-/-) mice. To account for the underrepresentation of Brg1 (+/-) ;Brm (-/-) mice, timed matings and blastocyst outgrowth assays demonstrated that ~50 % of these embryos failed to develop beyond the peri-implantation stage. These results challenge the idea that BRG1 is the exclusive catalytic subunit of SWI/SNF complexes in ES cells and suggest that BRM also interacts with the pluripotency transcription factors to facilitate self-renewal of the inner cell mass. In contrast to implantation, the Brm genotype did not influence an exencephaly phenotype that arises because of Brg1 haploinsufficiency during neural tube closure and that results in peri-natal lethality. Taken together, these results support the idea that BRG1 and BRM have overlapping functions for certain developmental processes but not others during embryogenesis.


Subject(s)
DNA Helicases/genetics , Embryonic Development/genetics , Gene Dosage , Nuclear Proteins/genetics , Transcription Factors/genetics , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Alleles , Animals , Blotting, Western , Catalytic Domain , Cells, Cultured , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Genotype , Mice , Nuclear Proteins/metabolism , Phenotype , Transcription Factors/metabolism
10.
J Cell Physiol ; 227(9): 3169-77, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22261928

ABSTRACT

Diet and energy metabolism affect gene expression, which influences human health and disease. Here, we discuss the role of epigenetics as a mechanistic link between energy metabolism and control of gene expression. A number of key energy metabolites including SAM, acetyl-CoA, NAD(+), and ATP serve as essential co-factors for many, perhaps most, epigenetic enzymes that regulate DNA methylation, posttranslational histone modifications, and nucleosome position. The relative abundance of these energy metabolites allows a cell to sense its energetic state. And as co-factors, energy metabolites act as rheostats to modulate the activity of epigenetic enzymes and upregulate/downregulate transcription as appropriate to maintain homeostasis.


Subject(s)
DNA Methylation/genetics , Diet , Energy Metabolism , Epigenesis, Genetic/genetics , Gene Expression Regulation , Acetyl Coenzyme A/metabolism , Adenosine Triphosphate/metabolism , Histones/genetics , Histones/metabolism , Humans , Metabolomics , NAD/metabolism , Protein Processing, Post-Translational/genetics , S-Adenosylmethionine/genetics , S-Adenosylmethionine/metabolism
11.
Nucleic Acids Res ; 38(20): 6906-19, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20571081

ABSTRACT

For DNA replication to occur, chromatin must be remodeled. Yet, we know very little about which proteins alter nucleosome occupancy at origins and replication forks and for what aspects of replication they are required. Here, we demonstrate that the BRG1 catalytic subunit of mammalian SWI/SNF-related complexes co-localizes with origin recognition complexes, GINS complexes, and proliferating cell nuclear antigen at sites of DNA replication on extended chromatin fibers. The specific pattern of BRG1 occupancy suggests it does not participate in origin selection but is involved in the firing of origins and the process of replication elongation. This latter function is confirmed by the fact that Brg1 mutant mouse embryos and RNAi knockdown cells exhibit a 50% reduction in replication fork progression rates, which is associated with decreased cell proliferation. This novel function of BRG1 is consistent with its requirement during embryogenesis and its role as a tumor suppressor to maintain genome stability and prevent cancer.


Subject(s)
DNA Helicases/physiology , DNA Replication , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , Cell Proliferation , Chromatin/chemistry , DNA Helicases/analysis , DNA Helicases/genetics , DNA-Binding Proteins/analysis , Embryonic Development , Erythroid Cells/metabolism , HeLa Cells , Humans , Mice , Mutation , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Phenotype , Transcription Factors/analysis , Transcription Factors/genetics
12.
Proc Natl Acad Sci U S A ; 106(7): 2259-64, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19171905

ABSTRACT

The dynamic packaging of DNA into chromatin is a fundamental step in the control of diverse nuclear processes. Whereas certain transcription factors and chromosomal components promote the formation of higher-order chromatin loops, the co-regulator machinery mediating loop assembly and disassembly is unknown. Using mice bearing a hypomorphic allele of the BRG1 chromatin remodeler, we demonstrate that the Brg1 mutation abrogated a cell type-specific loop between the beta-globin locus control region and the downstream beta major promoter, despite trans-acting factor occupancy at both sites. By contrast, distinct loops were insensitive to the Brg1 mutation. Molecular analysis with a conditional allele of GATA-1, a key regulator of hematopoiesis, in a novel cell-based system provided additional evidence that BRG1 functions early in chromatin domain activation to mediate looping. Although the paradigm in which chromatin remodelers induce nucleosome structural transitions is well established, our results demonstrating an essential role of BRG1 in the genesis of specific chromatin loops expands the repertoire of their functions.


Subject(s)
DNA Helicases/genetics , Mutation , Nuclear Proteins/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Alleles , Animals , CHO Cells , Chromatin/chemistry , Chromatin/metabolism , Cricetinae , Cricetulus , DNA/chemistry , GATA1 Transcription Factor/metabolism , Hematopoiesis , Mice , Nucleosomes/metabolism , beta-Globins/metabolism
13.
Nucleic Acids Res ; 37(18): 6019-27, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19696073

ABSTRACT

Alpha globin expression must be regulated properly to prevent the occurrence of alpha-thalassemias, yet many questions remain unanswered regarding the mechanism of transcriptional activation. Identifying factors that regulate chromatin structure of the endogenous alpha globin locus in developing erythroblasts will provide important mechanistic insight. Here, we demonstrate that the BRG1 catalytic subunit of SWI/SNF-related complexes co-immunoprecipitates with GATA-1 and EKLF in murine fetal liver cells in vivo and is recruited to the far-upstream major-regulatory element (MRE) and alpha2 promoter. Furthermore, based on our analysis of Brg1(null/ENU1) mutant mice, BRG1 regulates DNase I sensitivity, H3ac, and H3K4me2 but not CpG methylation at both sites. Most importantly, BRG1 is required for chromatin loop formation between the MRE and alpha2 promoter and for maximal RNA Polymerase II occupancy at the alpha2 promoter. Consequently, Brg1 mutants express alpha globin mRNA at only 5-10% of wild-type levels and die at mid-gestation. These data identify BRG1 as a chromatin-modifying factor required for nucleosome remodeling and transcriptional activation of the alpha globin locus. These data also demonstrate that chromatin looping between the MRE and alpha2 promoter is required as part of the transcriptional activation mechanism.


Subject(s)
Chromatin/chemistry , DNA Helicases/metabolism , Nuclear Proteins/metabolism , Nucleosomes/chemistry , Transcription Factors/metabolism , Transcriptional Activation , alpha-Globins/genetics , Animals , DNA Helicases/genetics , Deoxyribonuclease I , Epigenesis, Genetic , Mice , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Response Elements , Transcription Factors/genetics , alpha-Globins/metabolism
14.
Mol Cell Biol ; 27(12): 4551-65, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17438135

ABSTRACT

GATA factors orchestrate hematopoiesis via multistep transcriptional mechanisms, but the interrelationships and importance of individual steps are poorly understood. Using complementation analysis with GATA-1-null cells and mice containing a hypomorphic allele of the chromatin remodeler BRG1, we dissected the pathway from GATA-1 binding to cofactor recruitment, chromatin loop formation, and transcriptional activation. Analysis of GATA-1-mediated activation of the beta-globin locus, in which GATA-1 assembles dispersed complexes at the promoters and the distal locus control region (LCR), revealed molecular intermediates, including GATA-1-independent and GATA-1-containing LCR subcomplexes, both defective in promoting loop formation. An additional intermediate consisted of an apparently normal LCR complex and a promoter complex with reduced levels of total RNA polymerase II (Pol II) and Pol II phosphorylated at serine 5 of the carboxy-terminal domain. Reduced BRG1 activity solely compromised Pol II and serine 5-phosphorylated Pol II occupancy at the promoter, phenocopying the LCR-deleted mouse. These studies defined a hierarchical order of GATA-1-triggered events at a complex locus and establish a novel mechanism of long-range gene regulation.


Subject(s)
Cell Differentiation , Chromatin/metabolism , GATA1 Transcription Factor/physiology , Gene Expression Regulation , Hematopoietic System/physiology , Alleles , Animals , Cell Line , Chromatin/chemistry , Chromatin Immunoprecipitation , DNA Helicases/genetics , DNA Helicases/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/genetics , Gene Deletion , Globins/genetics , Locus Control Region , Mice , Mice, Knockout , Models, Genetic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Temperature , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Genes Environ ; 42: 21, 2020.
Article in English | MEDLINE | ID: mdl-32514323

ABSTRACT

BACKGROUND: Exogenous formaldehyde is classified by the IARC as a Category 1 known human carcinogen. Meanwhile, a significant amount of endogenous formaldehyde is produced in the human body; as such, formaldehyde-derived DNA and protein adducts have been detected in animals and humans in the absence of major exogenous formaldehyde exposure. However, the toxicological effects of endogenous formaldehyde on individuals with normal DNA damage repair functions are not well understood. In this study, we attempted to generate C57BL/6 mice deficient in both Adh5 and Aldh2, which encode two major enzymes that metabolize endogenous formaldehyde, in order to understand the effects of endogenous formaldehyde on mice with normal DNA repair function. RESULTS: Due to deficiencies in both ADH5 and ALDH2, few mice survived past post-natal day 21. In fact, the survival of pups within the first few days after birth was significantly decreased. Remarkably, two Aldh2 -/- /Adh5 -/- mice survived for 25 days after birth, and we measured their total body weight and organ weights. The body weight of Aldh2 -/- /Adh5 -/- mice decreased significantly by almost 37% compared to the Aldh2 -/- /Adh5 +/- and Aldh2 -/- /Adh5 +/+ mice of the same litter. In addition, the absolute weight of each organ was also significantly reduced. CONCLUSION: Mice deficient in both formaldehyde-metabolizing enzymes ADH5 and ALDH2 were found to develop partial synthetic lethality and mortality shortly after birth. This phenotype may be due to the accumulation of endogenous formaldehyde. No serious phenotype has been reported in people with dysfunctional, dominant-negative ALDH2*2 alleles, but it has been reported that they may be highly susceptible to osteoporosis and neurodegenerative diseases. It is important to further investigate these diseases in individuals with ALDH2*2 alleles, including an association with decreased metabolism, and thus accumulation, of formaldehyde.

16.
Vascul Pharmacol ; 133-134: 106777, 2020.
Article in English | MEDLINE | ID: mdl-32750408

ABSTRACT

Atherosclerosis is a systemic chronic inflammatory disease. Many antioxidants including alpha-lipoic acid (LA), a product of lipoic acid synthase (Lias), have proven to be effective for treatment of this disease. However, the question remains whether LA regulates the immune response as a protective mechanism against atherosclerosis. We initially investigated whether enhanced endogenous antioxidant can retard the development of atherosclerosis via immunomodulation. To explore the impact of enhanced endogenous antioxidant on the retardation of atherosclerosis via immune regulation, our laboratory has recently created a double mutant mouse model, using apolipoprotein E-deficient (Apoe-/-) mice crossbred with mice overexpressing lipoic acid synthase gene (LiasH/H), designated as LiasH/HApoe-/- mice. Their littermates, Lias+/+Apoe-/- mice, served as a control. Distinct redox environments between the two strains of mice have been established and they can be used to facilitate identification of antioxidant targets in the immune response. At 6 months of age, LiasH/HApoe-/- mice had profoundly decreased atherosclerotic lesion size in the aortic sinus compared to their Lias+/+Apoe-/- littermates, accompanied by significantly enhanced numbers of regulatory T cells (Tregs) and anti-oxidized LDL autoantibody in the vascular system, and reduced T cell infiltrates in aortic walls. Our results represent a novel exploration into an environment with increased endogenous antioxidant and its ability to alleviate atherosclerosis, likely through regulation of the immune response. These outcomes shed light on a new therapeutic strategy using antioxidants to lessen atherosclerosis.


Subject(s)
Aorta/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Plaque, Atherosclerotic , Sulfurtransferases/biosynthesis , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/immunology , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/immunology , Atherosclerosis/pathology , Autoantibodies/blood , Disease Models, Animal , Enzyme Induction , Lipoproteins, LDL/immunology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Oxidation-Reduction , Oxidative Stress , Sulfurtransferases/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
17.
Nat Microbiol ; 4(2): 375, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30607002

ABSTRACT

In the version of this News & Views originally published, owing to production error, the concentration scales given for butyrate circulates in the bloodstream and colon were incorrect. It was stated that "butyrate circulates at mM levels (as opposed to µM in colon)"; this should have read "butyrate circulates at µM levels (as opposed to mM in colon)". This error has now been corrected.

18.
Trends Endocrinol Metab ; 29(8): 529-531, 2018 08.
Article in English | MEDLINE | ID: mdl-29884327

ABSTRACT

In a recent Cell Reports article, Li et al. report that obesity is associated with altered fatty acid metabolism and DNA methylation in the colonic epithelium, which precede a tumor-prone gene-expression profile. Interestingly, obesity-associated methylation and transcriptome changes were reversed by weight loss, and the duration of weight loss correlated with the extent of restored gene expression. These findings have implications that are encouraging for weight loss and cancer prevention.


Subject(s)
Colorectal Neoplasms , Epigenesis, Genetic , Animals , DNA , Diet , Mice , Obesity/genetics , Transcriptome
19.
ACS Infect Dis ; 4(1): 46-52, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29094594

ABSTRACT

The intestinal epithelium provides a critical barrier that separates the gut microbiota from host tissues. Nonsteroidal anti-inflammatory drugs (NSAIDs) are efficacious analgesics and antipyretics and are among the most frequently used drugs worldwide. In addition to gastric damage, NSAIDs are toxic to the intestinal epithelium, causing erosions, perforations, and longitudinal ulcers in the gut. Here, we use a unique in vitro human primary small intestinal cell monolayer system to pinpoint the intestinal consequences of NSAID treatment. We found that physiologically relevant doses of the NSAID diclofenac (DCF) are cytotoxic because they uncouple mitochondrial oxidative phosphorylation and generate reactive oxygen species. We also find that DCF induces intestinal barrier permeability, facilitating the translocation of compounds from the luminal to the basolateral side of the intestinal epithelium. The results we outline here establish the utility of this novel platform, representative of the human small intestinal epithelium, to understand NSAID toxicity, which can be applied to study multiple aspects of gut barrier function including defense against infectious pathogens and host-microbiota interactions.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cell Membrane Permeability/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism
20.
Cell Mol Gastroenterol Hepatol ; 5(2): 113-130, 2018.
Article in English | MEDLINE | ID: mdl-29693040

ABSTRACT

BACKGROUND & AIMS: The successful culture of intestinal organoids has greatly enhanced our understanding of intestinal stem cell physiology and enabled the generation of novel intestinal disease models. Although of tremendous value, intestinal organoid culture systems have not yet fully recapitulated the anatomy or physiology of the in vivo intestinal epithelium. The aim of this work was to re-create an intestinal epithelium with a high density of polarized crypts that respond in a physiologic manner to addition of growth factors, metabolites, or cytokines to the basal or luminal tissue surface as occurs in vivo. METHODS: A self-renewing monolayer of human intestinal epithelium was cultured on a collagen scaffold microfabricated with an array of crypt-like invaginations. Placement of chemical factors in either the fluid reservoir below or above the cell-covered scaffolding created a gradient of that chemical across the growing epithelial tissue possessing the in vitro crypt structures. Crypt polarization (size of the stem/proliferative and differentiated cell zones) was assessed in response to gradients of growth factors, cytokines, and bacterial metabolites. RESULTS: Chemical gradients applied to the shaped human epithelium re-created the stem/proliferative and differentiated cell zones of the in vivo intestine. Short-chain fatty acids applied as a gradient from the luminal side confirmed long-standing hypotheses that butyrate diminished stem/progenitor cell proliferation and promoted differentiation into absorptive colonocytes. A gradient of interferon-γ and tumor necrosis factor-α significantly suppressed the stem/progenitor cell proliferation, altering crypt formation. CONCLUSIONS: The in vitro human colon crypt array accurately mimicked the architecture, luminal accessibility, tissue polarity, cell migration, and cellular responses of in vivo intestinal crypts.

SELECTION OF CITATIONS
SEARCH DETAIL