Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
N Engl J Med ; 387(4): 332-344, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35939579

ABSTRACT

BACKGROUND: Exome sequencing in hundreds of thousands of persons may enable the identification of rare protein-coding genetic variants associated with protection from human diseases like liver cirrhosis, providing a strategy for the discovery of new therapeutic targets. METHODS: We performed a multistage exome sequencing and genetic association analysis to identify genes in which rare protein-coding variants were associated with liver phenotypes. We conducted in vitro experiments to further characterize associations. RESULTS: The multistage analysis involved 542,904 persons with available data on liver aminotransferase levels, 24,944 patients with various types of liver disease, and 490,636 controls without liver disease. We found that rare coding variants in APOB, ABCB4, SLC30A10, and TM6SF2 were associated with increased aminotransferase levels and an increased risk of liver disease. We also found that variants in CIDEB, which encodes a structural protein found in hepatic lipid droplets, had a protective effect. The burden of rare predicted loss-of-function variants plus missense variants in CIDEB (combined carrier frequency, 0.7%) was associated with decreased alanine aminotransferase levels (beta per allele, -1.24 U per liter; 95% confidence interval [CI], -1.66 to -0.83; P = 4.8×10-9) and with 33% lower odds of liver disease of any cause (odds ratio per allele, 0.67; 95% CI, 0.57 to 0.79; P = 9.9×10-7). Rare coding variants in CIDEB were associated with a decreased risk of liver disease across different underlying causes and different degrees of severity, including cirrhosis of any cause (odds ratio per allele, 0.50; 95% CI, 0.36 to 0.70). Among 3599 patients who had undergone bariatric surgery, rare coding variants in CIDEB were associated with a decreased nonalcoholic fatty liver disease activity score (beta per allele in score units, -0.98; 95% CI, -1.54 to -0.41 [scores range from 0 to 8, with higher scores indicating more severe disease]). In human hepatoma cell lines challenged with oleate, CIDEB small interfering RNA knockdown prevented the buildup of large lipid droplets. CONCLUSIONS: Rare germline mutations in CIDEB conferred substantial protection from liver disease. (Funded by Regeneron Pharmaceuticals.).


Subject(s)
Apoptosis Regulatory Proteins , Germ-Line Mutation , Liver Diseases , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Genetic Predisposition to Disease/genetics , Genetic Predisposition to Disease/prevention & control , Humans , Liver/metabolism , Liver Diseases/genetics , Liver Diseases/metabolism , Liver Diseases/prevention & control , Transaminases/genetics , Exome Sequencing
2.
Mol Cell Proteomics ; 22(8): 100595, 2023 08.
Article in English | MEDLINE | ID: mdl-37328064

ABSTRACT

B4GALT1 encodes ß-1,4-galactosyltransferase 1, an enzyme that plays a major role in glycan synthesis in the Golgi apparatus by catalyzing the addition of terminal galactose. Studies increasingly suggest that B4GALT1 may be involved in the regulation of lipid metabolism pathways. Recently, we discovered a single-site missense variant Asn352Ser (N352S) in the functional domain of B4GALT1 in an Amish population, which decreases the level of LDL-cholesterol (LDL-c) as well as the protein levels of ApoB, fibrinogen, and IgG in the blood. To systematically evaluate the effects of this missense variant on protein glycosylation, expression, and secretion, we developed a nano-LC-MS/MS-based platform combined with TMT-labeling for in-depth quantitative proteomic and glycoproteomic analyses in the plasma of individuals homozygous for the B4GALT1 missense variant N352S versus non-carriers (n = 5 per genotype). A total of 488 secreted proteins in the plasma were identified and quantified, 34 of which showed significant fold changes in protein levels between N352S homozygotes and non-carriers. We determined N-glycosylation profiles from 370 glycosylation sites in 151 glycoproteins and identified ten proteins most significantly associated with decreased galactosylation and sialyation in B4GALT1 N352S homozygotes. These results further support that B4GALT1 N352S alters the glycosylation profiles of a variety of critical target proteins, thus governing the functions of these proteins in multiple pathways, such as those involved in lipid metabolism, coagulation, and the immune response.


Subject(s)
Galactosyltransferases , Proteomics , Humans , Amish/genetics , Galactosyltransferases/genetics , Galactosyltransferases/chemistry , Galactosyltransferases/metabolism , Glycosylation , Tandem Mass Spectrometry
3.
Nat Immunol ; 11(3): 207-15, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20081848

ABSTRACT

Hematopoietic stem cell (HSC) differentiation is regulated by cell-intrinsic and cell-extrinsic cues. In addition to transcriptional regulation, post-translational regulation may also control HSC differentiation. To test this hypothesis, we visualized the ubiquitin-regulated protein stability of a single transcription factor, c-Myc. The stability of c-Myc protein was indicative of HSC quiescence, and c-Myc protein abundance was controlled by the ubiquitin ligase Fbw7. Fine changes in the stability of c-Myc protein regulated the HSC gene-expression signature. Using whole-genome genomic approaches, we identified specific regulators of HSC function directly controlled by c-Myc binding; however, adult HSCs and embryonic stem cells sensed and interpreted c-Myc-regulated gene expression in distinct ways. Our studies show that a ubiquitin ligase-substrate pair can orchestrate the molecular program of HSC differentiation.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Ubiquitin-Protein Ligases/immunology , Animals , Cell Cycle/genetics , Cell Cycle/immunology , Cell Cycle Proteins/immunology , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Flow Cytometry , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myc/immunology
4.
J Pediatr ; 213: 137-142.e2, 2019 10.
Article in English | MEDLINE | ID: mdl-31327562

ABSTRACT

OBJECTIVE: To investigate whether the addition of the probiotic Lactobacillus rhamnosus GG (LGG) to the extensively hydrolyzed casein formula (EHCF) for cow's milk allergy (CMA) treatment could reduce the occurrence of functional gastrointestinal disorders (FGIDs). STUDY DESIGN: This cohort study included children with a positive history for CMA in the first year of life who were treated with EHCF alone or in combination with LGG and had evidence of immune tolerance acquisition to cow's milk for at least 12 months. FGID was diagnosed according to the Rome III diagnostic criteria by investigators unaware of previous treatment. A cohort of consecutive healthy children was also evaluated as a control population. RESULTS: A total of 330 subjects were included, 110 per cohort (EHCF, EHCF+LGG, and healthy controls). The rate of subjects with ≥1 FGID was significantly lower in the EHCF+LGG cohort compared with the EHCF cohort (40% vs 16.4%; P < .05). In the EHCF+LGG cohort, a lower incidence was observed for all components of the main study outcome. The prevalence of FGIDs in the healthy cohort was lower than that in the EHCF cohort and similar to that in the EHCF+LGG cohort. The incidence rate ratio of FGIDs for the EHCF+LGG cohort vs the EHCF cohort (0.40; 95% CI, 0.25-0.65; P < .001) was unmodified after correction for age at CMA diagnosis, breastfeeding, weaning time, and presence of a first-degree relative with an FGID. CONCLUSIONS: These results confirm the increased risk for developing FGIDs in children with CMA and suggest that EHCF+LGG could reduce this risk.


Subject(s)
Caseins/chemistry , Food, Formulated , Gastrointestinal Diseases/prevention & control , Lacticaseibacillus rhamnosus , Milk Hypersensitivity/diet therapy , Probiotics/administration & dosage , Animals , Cattle , Child , Child, Preschool , Diet , Female , Humans , Hydrolysis , Immune Tolerance , Male , Milk , Prevalence , Prospective Studies , Risk , Treatment Outcome
5.
Adv Exp Med Biol ; 1125: 57-68, 2019.
Article in English | MEDLINE | ID: mdl-30680644

ABSTRACT

The dramatic increase in food allergy prevalence and severity globally is demanding effective strategies. Food allergy derives from a defect in immune tolerance mechanisms. Immune tolerance is modulated by gut microbiota composition and function, and gut microbiota dysbiosis has been associated with the development of food allergy. Selected probiotic strains could act on immune tolerance mechanisms. The mechanisms are multiple and still not completely defined. Increasing evidence is providing useful information on the choice of optimal bacterial species/strains, dosage, and timing for intervention. The increased knowledge on the crucial role played by gut microbiota-derived metabolites, such as butyrate, is also opening the way to a post-biotic approach in the stimulation of immune tolerance.


Subject(s)
Food Hypersensitivity/therapy , Gastrointestinal Microbiome , Probiotics/therapeutic use , Dysbiosis/therapy , Humans , Immune Tolerance
6.
Int J Oncol ; 63(5)2023 11.
Article in English | MEDLINE | ID: mdl-37800623

ABSTRACT

T cell acute lymphoblastic leukemia (T­ALL), a neoplasm derived from T cell lineage­committed lymphoblasts, is characterized by genetic alterations that result in activation of oncogenic transcription factors and the NOTCH1 pathway activation. The NOTCH is a transmembrane receptor protein activated by γ­secretase. γ­secretase inhibitors (GSIs) are a NOTCH­targeted therapy for T­ALL. However, their clinical application has not been successful due to adverse events (primarily gastrointestinal toxicity), limited efficacy, and drug resistance caused by several mechanisms, including activation of the AKT/mTOR pathway. Nelfinavir is an human immunodeficiency virus 1 aspartic protease inhibitor and has been repurposed as an anticancer drug. It acts by inducing endoplasmic reticulum (ER) stress and inhibiting the AKT/mTOR pathway. Thus, it was hypothesized that nelfinavir might inhibit the NOTCH pathway via γ­secretase inhibition and blockade of aspartic protease presenilin, which would make nelfinavir effective against NOTCH­associated T­ALL. The present study assessed the efficacy of nelfinavir against T­ALL cells and investigated mechanisms of action in vitro and in preclinical treatment studies using a SCL­LMO1 transgenic mouse model. Nelfinavir blocks presenilin 1 processing and inhibits γ­secretase activity as well as the NOTCH1 pathway, thus suppressing T­ALL cell viability. Additionally, microarray analysis of nelfinavir­treated T­ALL cells showed that nelfinavir upregulated mRNA levels of CHAC1 (glutathione­specific γ­glutamylcyclotransferase 1, a negative regulator of NOTCH) and sestrin 2 (SESN2; a negative regulator of mTOR). As both factors are upregulated by ER stress, this confirmed that nelfinavir induced ER stress in T­ALL cells. Moreover, nelfinavir suppressed NOTCH1 mRNA expression in microarray analyses. These findings suggest that nelfinavir inhibited the NOTCH1 pathway by downregulating NOTCH1 mRNA expression, upregulating CHAC1 and suppressing γ­secretase via presenilin 1 inhibition and the mTOR pathway by upregulating SESN2 via ER stress induction. Further, nelfinavir exhibited therapeutic efficacy against T­ALL in an SCL­LMO1 transgenic mouse model. Collectively, these findings highlight the potential of nelfinavir as a novel therapeutic candidate for treatment of patients with T­ALL.


Subject(s)
Nelfinavir , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Mice , Animals , Nelfinavir/pharmacology , Nelfinavir/therapeutic use , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Presenilin-1 , Proto-Oncogene Proteins c-akt/metabolism , Amyloid Precursor Protein Secretases , TOR Serine-Threonine Kinases/metabolism , Enzyme Inhibitors , Transcription Factors , Mice, Transgenic , RNA, Messenger , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Cell Line, Tumor , Sestrins
7.
Commun Biol ; 5(1): 1051, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36192519

ABSTRACT

Glaucoma is a leading cause of blindness. Current glaucoma medications work by lowering intraocular pressure (IOP), a risk factor for glaucoma, but most treatments do not directly target the pathological changes leading to increased IOP, which can manifest as medication resistance as disease progresses. To identify physiological modulators of IOP, we performed genome- and exome-wide association analysis in >129,000 individuals with IOP measurements and extended these findings to an analysis of glaucoma risk. We report the identification and functional characterization of rare coding variants (including loss-of-function variants) in ANGPTL7 associated with reduction in IOP and glaucoma protection. We validated the human genetics findings in mice by establishing that Angptl7 knockout mice have lower (~2 mmHg) basal IOP compared to wild-type, with a trend towards lower IOP also in heterozygotes. Conversely, increasing murine Angptl7 levels via injection into mouse eyes increases the IOP. We also show that acute Angptl7 silencing in adult mice lowers the IOP (~2-4 mmHg), reproducing the observations in knockout mice. Collectively, our data suggest that ANGPTL7 is important for IOP homeostasis and is amenable to therapeutic modulation to help maintain a healthy IOP that can prevent onset or slow the progression of glaucoma.


Subject(s)
Glaucoma , Intraocular Pressure , Adult , Angiopoietin-Like Protein 7 , Angiopoietin-like Proteins/genetics , Animals , Blindness , Glaucoma/drug therapy , Glaucoma/genetics , Humans , Mice , Mice, Knockout
8.
Nat Commun ; 12(1): 5958, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34645820

ABSTRACT

Understanding the functional potential of the gut microbiome is of primary importance for the design of innovative strategies for allergy treatment and prevention. Here we report the gut microbiome features of 90 children affected by food (FA) or respiratory (RA) allergies and 30 age-matched, healthy controls (CT). We identify specific microbial signatures in the gut microbiome of allergic children, such as higher abundance of Ruminococcus gnavus and Faecalibacterium prausnitzii, and a depletion of Bifidobacterium longum, Bacteroides dorei, B. vulgatus and fiber-degrading taxa. The metagenome of allergic children shows a pro-inflammatory potential, with an enrichment of genes involved in the production of bacterial lipo-polysaccharides and urease. We demonstrate that specific gut microbiome signatures at baseline can be predictable of immune tolerance acquisition. Finally, a strain-level selection occurring in the gut microbiome of allergic subjects is identified. R. gnavus strains enriched in FA and RA showed lower ability to degrade fiber, and genes involved in the production of a pro-inflammatory polysaccharide. We demonstrate that a gut microbiome dysbiosis occurs in allergic children, with R. gnavus emerging as a main player in pediatric allergy. These findings may open new strategies in the development of innovative preventive and therapeutic approaches. Trial: NCT04750980.


Subject(s)
Allergens/immunology , Food Hypersensitivity/microbiology , Gastrointestinal Microbiome/immunology , Immune Tolerance , Respiratory Hypersensitivity/microbiology , Allergens/adverse effects , Animals , Bacteroides/isolation & purification , Bacteroides/metabolism , Bifidobacterium longum/isolation & purification , Bifidobacterium longum/metabolism , Case-Control Studies , Child , Child, Preschool , Clostridiales/isolation & purification , Clostridiales/metabolism , Dander/adverse effects , Dander/immunology , Eggs/adverse effects , Faecalibacterium prausnitzii/isolation & purification , Faecalibacterium prausnitzii/metabolism , Female , Food Hypersensitivity/etiology , Food Hypersensitivity/immunology , Humans , Lipopolysaccharides/biosynthesis , Male , Milk/adverse effects , Milk/immunology , Nuts/adverse effects , Nuts/immunology , Pollen/chemistry , Pollen/immunology , Prunus persica/chemistry , Prunus persica/immunology , Pyroglyphidae/chemistry , Pyroglyphidae/immunology , Respiratory Hypersensitivity/etiology , Respiratory Hypersensitivity/immunology , Urease/biosynthesis
9.
Science ; 374(6572): 1221-1227, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34855475

ABSTRACT

Increased blood levels of low-density lipoprotein cholesterol (LDL-C) and fibrinogen are independent risk factors for cardiovascular disease. We identified associations between an Amish-enriched missense variant (p.Asn352Ser) in a functional domain of beta-1,4-galactosyltransferase 1 (B4GALT1) and 13.9 milligrams per deciliter lower LDL-C (P = 4.1 × 10­19) and 29 milligrams per deciliter lower plasma fibrinogen (P = 1.3 × 10­5). B4GALT1 gene­based analysis in 544,955 subjects showed an association with decreased coronary artery disease (odds ratio = 0.64, P = 0.006). The mutant protein had 50% lower galactosyltransferase activity compared with the wild-type protein. N-linked glycan profiling of human serum found serine 352 allele to be associated with decreased galactosylation and sialylation of apolipoprotein B100, fibrinogen, immunoglobulin G, and transferrin. B4galt1 353Ser knock-in mice showed decreases in LDL-C and fibrinogen. Our findings suggest that targeted modulation of protein galactosylation may represent a therapeutic approach to decreasing cardiovascular disease.


Subject(s)
Cholesterol, LDL/blood , Fibrinogen/analysis , Galactosyltransferases/genetics , Mutation, Missense , Animals , Coronary Artery Disease/genetics , Coronary Artery Disease/prevention & control , Female , Galactose/metabolism , Galactosyltransferases/metabolism , Gene Knock-In Techniques , Gene Knockdown Techniques , Glycoproteins/blood , Glycosylation , Humans , Liver/enzymology , Male , Mice , N-Acetylneuraminic Acid/metabolism , Polysaccharides/blood , Whole Genome Sequencing
10.
Aliment Pharmacol Ther ; 51(1): 110-120, 2020 01.
Article in English | MEDLINE | ID: mdl-31797399

ABSTRACT

BACKGROUND: The pathogenesis of infant colic is poorly defined. Gut microbiota seems to be involved, supporting the potential therapeutic role of probiotics. AIMS: To assess the rate of infants with a reduction of ≥50% of mean daily crying duration after 28 days of intervention with the probiotic Bifidobacterium animalis subsp. lactis BB-12® (BB-12). Secondary outcomes were daily number of crying episodes, sleeping time, number of bowel movements and stool consistency. METHODS: Randomized controlled trial (RCT) on otherwise healthy exclusively breastfed infants with infant colic randomly allocated to receive BB-12 (1 × 109  CFU/day) or placebo for 28 days. Gut microbiota structure and butyrate, beta-defensin-2 (HBD-2), cathelicidin (LL-37), secretory IgA (sIgA) and faecal calprotectin levels were assessed. RESULTS: Eighty infants were randomised, 40/group. The rate of infants with reduction of ≥50% of mean daily crying duration was higher in infants treated with BB-12, starting from the end of 2nd week. No infant relapsed when treatment was stopped. The mean number of crying episodes decreased in both groups, but with a higher effect in BB-12 group (-4.7 ± 3.4 vs -2.3 ± 2.2, P < 0.05). Mean daily stool frequency decreased in both groups but the effect was significantly higher in the BB-12 group; stool consistency was similar between the two groups. An increase in Bifidobacterium abundance (with significant correlation with crying time reduction), butyrate and HBD-2, LL-37, sIgA levels associated with a decrease in faecal calprotectin level were observed in the BB-12 group. CONCLUSIONS: Supplementation with BB-12 is effective in managing infant colic. The effect could derive from immune and non-immune mechanisms associated with a modulation of gut microbiota structure and function.


Subject(s)
Bifidobacterium animalis , Colic/diet therapy , Probiotics/therapeutic use , Breast Feeding , Colic/microbiology , Crying , Defecation , Double-Blind Method , Feces/microbiology , Female , Gastrointestinal Microbiome/physiology , Humans , Infant , Infant Care/methods , Male , Placebos , Treatment Outcome
11.
Blood Cancer Discov ; 1(2): 178-197, 2020 09.
Article in English | MEDLINE | ID: mdl-32924017

ABSTRACT

Notch activation is highly prevalent among cancers, in particular T-cell acute lymphoblastic leukemia (T-ALL). However, the use of pan-Notch inhibitors to treat cancers has been hampered by adverse effects, particularly intestinal toxicities. To circumvent this barrier in T-ALL, we aimed to inhibit ETS1, a developmentally important T-cell transcription factor previously shown to co-bind Notch response elements. Using complementary genetic approaches in mouse models, we show that ablation of Ets1 leads to strong Notch-mediated suppressive effects on T-cell development and leukemogenesis, but milder intestinal effects than pan-Notch inhibitors. Mechanistically, genome-wide chromatin profiling studies demonstrate that Ets1 inactivation impairs recruitment of multiple Notch-associated factors and Notch-dependent activation of transcriptional elements controlling major Notch-driven oncogenic effector pathways. These results uncover previously unrecognized hierarchical heterogeneity of Notch-controlled genes and points to Ets1-mediated enucleation of Notch-Rbpj transcriptional complexes as a target for developing specific anti-Notch therapies in T-ALL that circumvent the barriers of pan-Notch inhibition.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Leukemia, T-Cell , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Proto-Oncogene Protein c-ets-1 , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/drug effects , Leukemia, T-Cell/drug therapy , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Receptor, Notch1/antagonists & inhibitors , Signal Transduction/physiology
12.
Front Immunol ; 10: 191, 2019.
Article in English | MEDLINE | ID: mdl-30828329

ABSTRACT

The dramatic increase in food allergy prevalence and severity globally requires effective strategies. Food allergy derives from a defect in immune tolerance mechanisms. Immune tolerance is modulated by gut microbiota function and structure, and microbiome alterations (dysbiosis) have a pivotal role in the development of food allergy. Environmental factors, including a low-fiber/high-fat diet, cesarean delivery, antiseptic agents, lack of breastfeeding, and drugs can induce gut microbiome dysbiosis, and have been associated with food allergy. New experimental tools and technologies have provided information regarding the role of metabolites generated from dietary nutrients and selected probiotic strains that could act on immune tolerance mechanisms. The mechanisms are multiple and still not completely defined. Increasing evidence has provided useful information on optimal bacterial species/strains, dosage, and timing for intervention. The increased knowledge of the crucial role played by nutrients and gut microbiota-derived metabolites is opening the way to a post-biotic approach in the stimulation of immune tolerance through epigenetic regulation. This review focused on the potential role of gut microbiome as the target for innovative strategies against food allergy.


Subject(s)
Food Hypersensitivity/immunology , Food Hypersensitivity/therapy , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Age Factors , Animals , Diet , Disease Management , Disease Susceptibility , Dysbiosis/immunology , Fatty Acids/metabolism , Food Hypersensitivity/metabolism , Humans , Immune Tolerance , Probiotics
13.
Sci Rep ; 8(1): 12500, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30131575

ABSTRACT

Cow's milk allergy (CMA) is one of the earliest and most common food allergy and can be elicited by both IgE- or non-IgE-mediated mechanism. We previously described dysbiosis in children with IgE-mediated CMA and the effect of dietary treatment with extensively hydrolyzed casein formula (EHCF) alone or in combination with the probiotic Lactobacillus rhamnosus GG (LGG). On the contrary, the gut microbiota in non-IgE-mediated CMA remains uncharacterized. In this study we evaluated gut microbiota composition and fecal butyrate levels in children affected by non-IgE-mediated CMA. We found a gut microbiota dysbiosis in non-IgE-mediated CMA, driven by an enrichment of Bacteroides and Alistipes. Comparing these results with those previously obtained in children with IgE-mediated CMA, we demonstrated overlapping signatures in the gut microbiota dysbiosis of non-IgE-mediated and IgE-mediated CMA children, characterized by a progressive increase in Bacteroides from healthy to IgE-mediated CMA patients. EHCF containg LGG was more strongly associated with an effect on dysbiosis and on butyrate production if compared to what observed in children treated with EHCF alone. If longitudinal cohort studies in children with CMA will confirm these results, gut microbiota dysbiosis could be a relevant target for innovative therapeutic strategies in children with non-IgE-mediated CMA.


Subject(s)
Bacteria/classification , Butyrates/analysis , Dysbiosis/diagnosis , Milk Hypersensitivity/microbiology , Sequence Analysis, DNA/methods , Animals , Bacteria/genetics , Bacteria/isolation & purification , Bacteroides/classification , Bacteroides/genetics , Bacteroides/isolation & purification , Child, Preschool , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Dysbiosis/etiology , Feces/chemistry , Gastrointestinal Microbiome , Humans , Infant , Longitudinal Studies , Milk Hypersensitivity/metabolism , RNA, Ribosomal, 16S/genetics
15.
Nat Med ; 18(3): 436-40, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22366949

ABSTRACT

The TLX1 and TLX3 transcription factor oncogenes have a key role in the pathogenesis of T cell acute lymphoblastic leukemia (T-ALL). Here we used reverse engineering of global transcriptional networks to decipher the oncogenic regulatory circuit controlled by TLX1 and TLX3. This systems biology analysis defined T cell leukemia homeobox 1 (TLX1) and TLX3 as master regulators of an oncogenic transcriptional circuit governing T-ALL. Notably, a network structure analysis of this hierarchical network identified RUNX1 as a key mediator of the T-ALL induced by TLX1 and TLX3 and predicted a tumor-suppressor role for RUNX1 in T cell transformation. Consistent with these results, we identified recurrent somatic loss-of-function mutations in RUNX1 in human T-ALL. Overall, these results place TLX1 and TLX3 at the top of an oncogenic transcriptional network controlling leukemia development, show the power of network analyses to identify key elements in the regulatory circuits governing human cancer and identify RUNX1 as a tumor-suppressor gene in T-ALL.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Regulatory Networks/genetics , Homeodomain Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Homeodomain Proteins/genetics , Humans , Mutation , Oligonucleotide Array Sequence Analysis , Oncogenes/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Conformation , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
16.
J Exp Med ; 208(13): 2571-9, 2011 Dec 19.
Article in English | MEDLINE | ID: mdl-22162831

ABSTRACT

Early immature T cell acute lymphoblastic leukemias (T-ALLs) account for ~5-10% of pediatric T-ALLs and are associated with poor prognosis. However, the genetic defects that drive the biology of these tumors remain largely unknown. In this study, analysis of microarray gene expression signatures in adult T-ALL demonstrated a high prevalence of early immature leukemias and revealed a close relationship between these tumors and myeloid leukemias. Many adult immature T-ALLs harbored mutations in myeloid-specific oncogenes and tumor suppressors including IDH1, IDH2, DNMT3A, FLT3, and NRAS. Moreover, we identified ETV6 mutations as a novel genetic lesion uniquely present in immature adult T-ALL. Our results demonstrate that early immature adult T-ALL represents a heterogeneous category of leukemias characterized by the presence of overlapping myeloid and T-ALL characteristics, and highlight the potential role of ETV6 mutations in these tumors.


Subject(s)
Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Adult , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic/genetics , Genes, Tumor Suppressor , Humans , Infant , Male , Oligonucleotide Array Sequence Analysis , ETS Translocation Variant 6 Protein
17.
Genome Res ; 18(6): 939-48, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18441228

ABSTRACT

Genome-wide identification of bona-fide targets of transcription factors in mammalian cells is still a challenge. We present a novel integrated computational and experimental approach to identify direct targets of a transcription factor. This consists of measuring time-course (dynamic) gene expression profiles upon perturbation of the transcription factor under study, and in applying a novel "reverse-engineering" algorithm (TSNI) to rank genes according to their probability of being direct targets. Using primary keratinocytes as a model system, we identified novel transcriptional target genes of TRP63, a crucial regulator of skin development. TSNI-predicted TRP63 target genes were validated by Trp63 knockdown and by ChIP-chip to identify TRP63-bound regions in vivo. Our study revealed that short sampling times, in the order of minutes, are needed to capture the dynamics of gene expression in mammalian cells. We show that TRP63 transiently regulates a subset of its direct targets, thus highlighting the importance of considering temporal dynamics when identifying transcriptional targets. Using this approach, we uncovered a previously unsuspected transient regulation of the AP-1 complex by TRP63 through direct regulation of a subset of AP-1 components. The integrated experimental and computational approach described here is readily applicable to other transcription factors in mammalian systems and is complementary to genome-wide identification of transcription-factor binding sites.


Subject(s)
Algorithms , Gene Expression Profiling , Gene Regulatory Networks , Phosphoproteins/metabolism , Trans-Activators/metabolism , Animals , Binding Sites , Cells, Cultured , Chromatin Immunoprecipitation , Computational Biology , Humans , Keratinocytes/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Transcription Factor AP-1/metabolism
18.
J Invest Dermatol ; 128(7): 1676-85, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18256694

ABSTRACT

p63 and p73 are highly homologous members of the p53 family that originated by gene duplication at the invertebrate-to-vertebrate transition. We characterize here a previously unreported gene, Transformation-related protein 63 regulated (Tprg), located upstream of the p63 gene in the vertebrate genome, with striking similarity to Transformation related protein 63 regulated like (Tprgl), an uncharacterized gene located upstream of p73, suggesting that p63/Tprg and p73/Tprgl are embedded in a paralogue region originated from a single duplication event. Tprg is predominantly expressed in the epithelial compartment of the skin, more abundantly in differentiated cells. Consistent with its relative higher expression in differentiated keratinocytes, finely tuned p63 expression levels are required for optimal Tprg expression in primary keratinocytes. p63 is essential for Tprg expression as shown in p63-knockdown keratinocytes; however, high levels of p63 result in Tprg downregulation. p63 directly binds in vivo to a canonical p63-binding site in an evolutionary conserved genomic region located in Tprg intron 4. This genomic region is sufficient to function as a p63-inducible enhancer in promoter studies. Thus, we demonstrate that the Tprg gene is predominantly expressed in skin, is physically associated with the p63 gene during evolution, and directly regulated by p63 through a long-distance enhancer located within the Tprg locus.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Phosphoproteins/physiology , Skin/metabolism , Trans-Activators/physiology , Tumor Suppressor Proteins/physiology , Animals , Calcium/physiology , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/genetics , Humans , Keratinocytes/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phosphoproteins/genetics , Signal Transduction , Skin/embryology , Trans-Activators/genetics , Transcription Factors , Tumor Protein p73 , Tumor Suppressor Proteins/genetics
19.
Bioinformatics ; 22(7): 815-22, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16418235

ABSTRACT

MOTIVATION: Time series expression experiments are an increasingly popular method for studying a wide range of biological systems. Here we developed an algorithm that can infer the local network of gene-gene interactions surrounding a gene of interest. This is achieved by a perturbation of the gene of interest and subsequently measuring the gene expression profiles at multiple time points. We applied this algorithm to computer simulated data and to experimental data on a nine gene network in Escherichia coli. RESULTS: In this paper we show that it is possible to recover the gene regulatory network from a time series data of gene expression following a perturbation to the cell. We show this both on simulated data and on a nine gene subnetwork part of the DNA-damage response pathway (SOS pathway) in the bacteria E. coli. CONTACT: dibernardo@tigem.it SUPLEMENTARY INFORMATION: Supplementary data are available at http://dibernado.tigem.it


Subject(s)
Algorithms , Gene Expression Profiling/methods , Bayes Theorem , Cell Physiological Phenomena , Computer Simulation , Escherichia coli/genetics , Escherichia coli/metabolism , Models, Biological , Models, Statistical , Time Factors
20.
Genes Dev ; 20(8): 1028-42, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16618808

ABSTRACT

Notch signaling promotes commitment of keratinocytes to differentiation and suppresses tumorigenesis. p63, a p53 family member, has been implicated in establishment of the keratinocyte cell fate and/or maintenance of epithelial self-renewal. Here we show that p63 expression is suppressed by Notch1 activation in both mouse and human keratinocytes through a mechanism independent of cell cycle withdrawal and requiring down-modulation of selected interferon-responsive genes, including IRF7 and/or IRF3. In turn, elevated p63 expression counteracts the ability of Notch1 to restrict growth and promote differentiation. p63 functions as a selective modulator of Notch1-dependent transcription and function, with the Hes-1 gene as one of its direct negative targets. Thus, a complex cross-talk between Notch and p63 is involved in the balance between keratinocyte self-renewal and differentiation.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/physiology , Keratinocytes/cytology , Receptor, Notch1/physiology , Trans-Activators/physiology , Tumor Suppressor Proteins/physiology , Animals , Base Sequence , DNA Primers , Humans , Mice , Promoter Regions, Genetic , RNA, Small Interfering , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL