Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Commun Biol ; 5(1): 992, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36127469

ABSTRACT

Rhabdomyosarcoma, the most common pediatric sarcoma, has no effective treatment for the pleomorphic subtype. Still, what triggers transformation into this aggressive phenotype remains poorly understood. Here we used Ptch1+/-/ETV7TG/+/- mice with enhanced incidence of rhabdomyosarcoma to generate a model of pleomorphic rhabdomyosarcoma driven by haploinsufficiency of the lysosomal sialidase neuraminidase 1. These tumors share mostly features of embryonal and some of alveolar rhabdomyosarcoma. Mechanistically, we show that the transforming pathway is increased lysosomal exocytosis downstream of reduced neuraminidase 1, exemplified by the redistribution of the lysosomal associated membrane protein 1 at the plasma membrane of tumor and stromal cells. Here we exploit this unique feature for single cell analysis and define heterogeneous populations of exocytic, only partially differentiated cells that force tumors to pleomorphism and promote a fibrotic microenvironment. These data together with the identification of an adipogenic signature shared by human rhabdomyosarcoma, and likely fueling the tumor's metabolism, make this model of pleomorphic rhabdomyosarcoma ideal for diagnostic and therapeutic studies.


Subject(s)
Neuraminidase , Rhabdomyosarcoma , Animals , Haploinsufficiency , Humans , Lysosomal-Associated Membrane Protein 1 , Lysosomes/metabolism , Mice , Neuraminidase/genetics , Neuraminidase/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Tumor Microenvironment
2.
Biochim Biophys Acta Rev Cancer ; 1876(1): 188571, 2021 08.
Article in English | MEDLINE | ID: mdl-34051287

ABSTRACT

Pediatric cancer is a rare disease with a distinct etiology and mutational landscape compared with adult cancer. Multi-omics profiling of retrospective and prospective cohorts coupled with innovative computational analysis have been instrumental in uncovering mechanisms of tumorigenesis and drug resistance that are now informing pediatric cancer clinical therapy. In this review we present the major data resources of pediatric cancer and actionable insights into pediatric cancer etiology stemming from the identification of oncogenic gene fusions, mutational signature analysis, systems biology, cancer predisposition and survivorship studies - that have led to improved clinical diagnosis, discovery of new drug-targets, pharmacological therapy, and screening for genetic predisposition. Ultimately, integration of large-scale omics datasets generated through international collaboration is required to maximize the power of data-driven approaches to advance pediatric cancer research informing clinical therapy.


Subject(s)
Biomarkers, Tumor/genetics , Databases, Genetic , Genomics , Machine Learning , Medical Oncology , Mutation , Neoplasms/genetics , Pediatrics , Age of Onset , Data Mining , Genetic Predisposition to Disease , Humans , Neoplasms/epidemiology , Neoplasms/therapy , Precision Medicine , Transcriptome
3.
Cell Rep ; 32(3): 107922, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32698014

ABSTRACT

Spatiotemporal control of Wnt/ß-catenin signaling is critical for organism development and homeostasis. The poly-(ADP)-ribose polymerase Tankyrase (TNKS1) promotes Wnt/ß-catenin signaling through PARylation-mediated degradation of AXIN1, a component of the ß-catenin destruction complex. Although Wnt/ß-catenin is a niche-restricted signaling program, tissue-specific factors that regulate TNKS1 are not known. Here, we report prostate-associated gene 4 (PAGE4) as a tissue-specific TNKS1 inhibitor that robustly represses canonical Wnt/ß-catenin signaling in human cells, zebrafish, and mice. Structural and biochemical studies reveal that PAGE4 acts as an optimal substrate decoy that potently hijacks substrate binding sites on TNKS1 to prevent AXIN1 PARylation and degradation. Consistently, transgenic expression of PAGE4 in mice phenocopies TNKS1 knockout. Physiologically, PAGE4 is selectively expressed in stromal prostate fibroblasts and functions to establish a proper Wnt/ß-catenin signaling niche through suppression of autocrine signaling. Our findings reveal a non-canonical mechanism for TNKS1 inhibition that functions to establish tissue-specific control of the Wnt/ß-catenin pathway.


Subject(s)
Antigens, Neoplasm/metabolism , Organ Specificity , Tankyrases/antagonists & inhibitors , Wnt Signaling Pathway , Amino Acid Motifs , Amino Acid Sequence , Animals , Antigens, Neoplasm/chemistry , Axin Protein , Fibroblasts/metabolism , HEK293 Cells , Humans , Male , Mice, Knockout , Models, Biological , Poly ADP Ribosylation , Prostate/metabolism , Protein Domains , Proteolysis , Stromal Cells/metabolism , Substrate Specificity , Tankyrases/chemistry , Tankyrases/metabolism , Ubiquitination , Zebrafish
4.
Cancer Cell ; 36(5): 512-527.e9, 2019 11 11.
Article in English | MEDLINE | ID: mdl-31631027

ABSTRACT

ATRX alterations occur at high frequency in neuroblastoma of adolescents and young adults. Particularly intriguing are the large N-terminal deletions of ATRX (Alpha Thalassemia/Mental Retardation, X-linked) that generate in-frame fusion (IFF) proteins devoid of key chromatin interaction domains, while retaining the SWI/SNF-like helicase region. We demonstrate that ATRX IFF proteins are redistributed from H3K9me3-enriched chromatin to promoters of active genes and identify REST as an ATRX IFF target whose activation promotes silencing of neuronal differentiation genes. We further show that ATRX IFF cells display sensitivity to EZH2 inhibitors, due to derepression of neurogenesis genes, including a subset of REST targets. Taken together, we demonstrate that ATRX structural alterations are not loss-of-function and put forward EZH2 inhibitors as a potential therapy for ATRX IFF neuroblastoma.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Neuroblastoma/drug therapy , Repressor Proteins/genetics , X-linked Nuclear Protein/genetics , Animals , Base Sequence/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Female , Histones/metabolism , Humans , Male , Mice , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/surgery , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Promoter Regions, Genetic , Protein Domains/genetics , Sequence Deletion , X-linked Nuclear Protein/metabolism , Xenograft Model Antitumor Assays
5.
PLoS One ; 14(2): e0211244, 2019.
Article in English | MEDLINE | ID: mdl-30807587

ABSTRACT

Expulsion of parasitic gastrointestinal nematodes requires diverse effector mechanisms coordinated by a Th2-type response. The evolutionarily conserved JmjC protein; Myc Induced Nuclear Antigen (Mina) has been shown to repress IL4, a key Th2 cytokine, suggesting Mina may negatively regulate nematode expulsion. Here we report that expulsion of the parasitic nematode Trichuris muris was indeed accelerated in Mina deficient mice. Unexpectedly, this was associated not with an elevated Th2- but rather an impaired Th1-type response. Further reciprocal bone marrow chimera and conditional KO experiments demonstrated that retarded parasite expulsion and a normal Th1-type response both required Mina in intestinal epithelial cells (IECs). Transcriptional profiling experiments in IECs revealed anti-microbial α-defensin peptides to be the major target of Mina-dependent retention of worms in infected mice. In vitro exposure to recombinant α-defensin peptides caused cytotoxic damage to whipworms. These results identify a latent IEC-intrinsic anthelmintic pathway actively constrained by Mina and point to α-defensins as important effectors that together with Mina may be attractive therapeutic targets for the control of nematode infection.


Subject(s)
Epithelial Cells/metabolism , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Trichuris/immunology , Animals , Cytokines/analysis , Epithelial Cells/cytology , Intestines/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/deficiency , Nuclear Proteins/deficiency , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism , Transcriptome , Trichuriasis/drug therapy , Trichuriasis/immunology , Trichuriasis/pathology , Trichuris/drug effects , Trichuris/pathogenicity , alpha-Defensins/genetics , alpha-Defensins/metabolism
6.
Neoplasia ; 18(3): 172-84, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26992918

ABSTRACT

The current paradigm of pancreatic neoplastic transformation proposes an initial step whereby acinar cells convert into acinar-to-ductal metaplasias, followed by progression of these lesions into neoplasias under sustained oncogenic activity and inflammation. Understanding the molecular mechanisms driving these processes is crucial to the early diagnostic and prevention of pancreatic cancer. Emerging evidence indicates that transcription factors that control exocrine pancreatic development could have either, protective or facilitating roles in the formation of preneoplasias and neoplasias in the pancreas. We previously identified that the homeodomain transcription factor Prox1 is a novel regulator of mouse exocrine pancreas development. Here we investigated whether Prox1 function participates in early neoplastic transformation using in vivo, in vitro and in silico approaches. We found that Prox1 expression is transiently re-activated in acinar cells undergoing dedifferentiation and acinar-to-ductal metaplastic conversion. In contrast, Prox1 expression is largely absent in neoplasias and tumors in the pancreas of mice and humans. We also uncovered that Prox1-heterozygosis markedly increases the formation of acinar-to-ductal-metaplasias and early neoplasias, and enhances features associated with inflammation, in mouse pancreatic tissues expressing oncogenic Kras. Furthermore, we discovered that Prox1-heterozygosis increases tissue damage and delays recovery from inflammation in pancreata of mice injected with caerulein. These results are the first demonstration that Prox1 activity protects pancreatic cells from acute tissue damage and early neoplastic transformation. Additional data in our study indicate that this novel role of Prox1 involves suppression of pathways associated with inflammatory responses and cell invasiveness.


Subject(s)
Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Proteins/genetics , Acinar Cells/pathology , Animals , Cell Transformation, Neoplastic/pathology , Ceruletide/administration & dosage , Heterozygote , Homeodomain Proteins/biosynthesis , Humans , Inflammation/genetics , Inflammation/pathology , Metaplasia/genetics , Metaplasia/pathology , Mice , Pancreas/pathology , Pancreatic Neoplasms/pathology , Tumor Suppressor Proteins/biosynthesis
7.
Biochemistry ; 55(9): 1332-45, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26866573

ABSTRACT

Proline-rich tyrosine kinase 2 (Pyk2) is a nonreceptor tyrosine kinase and belongs to the focal adhesion kinase (FAK) family. Like FAK, the C-terminal focal adhesion-targeting (FAT) domain of Pyk2 binds to paxillin, a scaffold protein in focal adhesions; however, the interaction between the FAT domain of Pyk2 and paxillin is dynamic and unstable. Leupaxin is another member in the paxillin family and was suggested to be the native binding partner of Pyk2; Pyk2 gene expression is strongly correlated with that of leupaxin in many tissues including primary breast cancer. Here, we report that leupaxin interacts with Pyk2-FAT. Leupaxin has four leucine-aspartate (LD) motifs. The first and third LD motifs of leupaxin preferably target the two LD-binding sites on the Pyk2-FAT domain, respectively. Moreover, the full-length leupaxin binds to Pyk2-FAT as a stable one-to-one complex. Together, we propose that there is an underlying selectivity between leupaxin and paxillin for Pyk2, which may influence the differing behavior of the two proteins at focal adhesion sites.


Subject(s)
Aspartic Acid/chemistry , Cell Adhesion Molecules/chemistry , Focal Adhesion Kinase 2/chemistry , Focal Adhesions/chemistry , Leucine/chemistry , Phosphoproteins/chemistry , Aspartic Acid/metabolism , Cell Adhesion Molecules/metabolism , Crystallization , Focal Adhesion Kinase 2/metabolism , Focal Adhesions/metabolism , Humans , Leucine/metabolism , Phosphoproteins/metabolism , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary/physiology
8.
Immunity ; 41(5): 853-65, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25517617

ABSTRACT

The molecular mechanisms that regulate the rapid transcriptional changes that occur during cytotoxic T lymphocyte (CTL) proliferation and differentiation in response to infection are poorly understood. We have utilized ChIP-seq to assess histone H3 methylation dynamics within naive, effector, and memory virus-specific T cells isolated directly ex vivo after influenza A virus infection. Our results show that within naive T cells, codeposition of the permissive H3K4me3 and repressive H3K27me3 modifications is a signature of gene loci associated with gene transcription, replication, and cellular differentiation. Upon differentiation into effector and/or memory CTLs, the majority of these gene loci lose repressive H3K27me3 while retaining the permissive H3K4me3 modification. In contrast, immune-related effector gene promoters within naive T cells lacked the permissive H3K4me3 modification, with acquisition of this modification occurring upon differentiation into effector/memory CTLs. Thus, coordinate transcriptional regulation of CTL genes with related functions is achieved via distinct epigenetic mechanisms.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic/immunology , Histones/genetics , Influenza A virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Cell Proliferation , DNA Methylation/genetics , Immunologic Memory , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Protein Processing, Post-Translational , T-Lymphocytes, Cytotoxic/cytology , Transcription, Genetic/immunology
9.
Proc Natl Acad Sci U S A ; 109(50): 20332-7, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23185025

ABSTRACT

Disentangling the effects of climate change and anthropogenic activities on the environment is a major challenge in paleoenvironmental research. Here, we used fecal sterols and other biogeochemical compounds in lake sediments from northern Norway to identify both natural and anthropogenic signals of environmental change during the late Holocene. The area was first occupied by humans and their grazing animals at ∼2,250 ± 75 calendar years before 1950 AD (calendar years before present). The arrival of humans is indicated by an abrupt increase in coprostanol (and its epimer epicoprostanol) in the sediments and an associated increase in 5ß-stigmastanol (and 5ß-epistigmastanol), which resulted from human and animal feces washing into the lake. Human settlement was accompanied by an abrupt increase in landscape fires (indicated by the rise in pyrolytic polycyclic aromatic hydrocarbons) and a decline in woodland (registered by a change in n-alkane chain lengths from leaf waxes), accelerating a process that began earlier in the Holocene. Human activity and associated landscape changes in the region over the last two millennia were mainly driven by summer temperatures, as indicated by independent tree-ring reconstructions, although there were periods when socioeconomic factors played an equally important role. In this study, fecal sterols in lake sediments have been used to provide a record of human occupancy through time. This approach may be useful in many archeological studies, both to confirm the presence of humans and grazing animals, and to distinguish between anthropogenic and natural factors that have influenced the environment in the past.


Subject(s)
Climate Change/history , Geologic Sediments/analysis , Agriculture/history , Animals , Archaeology , Environment , Feces/chemistry , History, Ancient , Humans , Norway , Sterols/analysis
10.
J Virol ; 81(19): 10292-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17652405

ABSTRACT

Avian influenza viruses have adapted to human hosts, causing pandemics in humans. The key host-specific amino acid mutations required for an avian influenza virus to function in humans are unknown. Through multiple-sequence alignment and statistical testing of each aligned amino acid, we identified markers that discriminate human influenza viruses from avian influenza viruses. We applied strict thresholds to select only markers which are highly preserved in human influenza virus isolates over time. We found that a subset of these persistent host markers exist in all human pandemic influenza virus sequences from 1918, 1957, and 1968, while others are acquired as the virus becomes a seasonal influenza virus. We also show that human H5N1 influenza viruses are significantly more likely to contain the amino acid predominant in human strains for a few persistent host markers than avian H5N1 influenza viruses. This sporadic enrichment of amino acids present in human-hosted viruses may indicate that some H5N1 viruses have made modest adaptations to their new hosts in the recent past. The markers reported here should be useful in monitoring potential pandemic influenza viruses.


Subject(s)
Birds/virology , Disease Outbreaks , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/virology , Influenza, Human/virology , Amino Acid Substitution/genetics , Animals , Evolution, Molecular , Genetic Markers , Humans , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza in Birds/epidemiology , Influenza, Human/epidemiology , Mutagenesis , Seasons , Sequence Alignment , Sequence Analysis, Protein , Sequence Analysis, RNA , Viral Proteins/genetics
11.
Science ; 311(5767): 1576-80, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16439620

ABSTRACT

The spread of H5N1 avian influenza viruses (AIVs) from China to Europe has raised global concern about their potential to infect humans and cause a pandemic. In spite of their substantial threat to human health, remarkably little AIV whole-genome information is available. We report here a preliminary analysis of the first large-scale sequencing of AIVs, including 2196 AIV genes and 169 complete genomes. We combine this new information with public AIV data to identify new gene alleles, persistent genotypes, compensatory mutations, and a potential virulence determinant.


Subject(s)
Genes, Viral , Influenza A Virus, H5N1 Subtype/genetics , Influenza A virus/genetics , Viral Nonstructural Proteins/chemistry , Virulence Factors/chemistry , Animals , Birds/virology , Computational Biology , Genome, Viral , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H2N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N8 Subtype/genetics , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N2 Subtype/genetics , Influenza A Virus, H7N7 Subtype/genetics , Influenza A Virus, H9N2 Subtype/genetics , Influenza A virus/chemistry , Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Influenza in Birds/virology , Influenza, Human/virology , Molecular Sequence Data , Mutation , Phylogeny , RNA, Viral/genetics , Reassortant Viruses/genetics , Sequence Analysis, DNA , Viral Nonstructural Proteins/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Virulence Factors/genetics
12.
J Biomol Tech ; 16(2): 143-53, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16030321

ABSTRACT

Improvements in the quality of gene expression data were investigated based on a database consisting of 5168 oligonucleotide microarrays collected over 3 years. The database includes diverse treatments of human and mouse samples collected from multiple laboratories. The array designs and algorithms used to capture the data have also changed over the 3 years of data collection. All hybridizations and labeling were conducted in the Hartwell Center for Bioinformatics and Biotechnology at St. Jude's Children's Research Hospital. Quality metrics for each human and mouse array were collected and analyzed. Statistical tests, such as ANOVA and linear regression, were applied to test for the effects of array design, algorithm, and time. The quality metrics tested were average background, actin 3'/5' ratio, Bio B signal, percent present, and scale factor. ANOVA results indicate that both recent algorithms and chip designs significantly correlate with improvements in Bio B, scale factor, percent present, and average background. Significant quality improvements correlated with new chip designs, algorithms, and their interaction. In addition, within one chip type analyzed by the same algorithm significant improving trends were still observed. Scale factor, percent present, and average background significantly improved over time for U133A arrays analyzed by the Affymetrix MicroArray Suite 5.0 algorithm according to linear regression. Proportionally fewer outlier arrays (those with less than 25% present calls) were seen over time. Also, high throughput periods did not increase the proportion of outliers, indicating that laboratory monitoring of quality is successfully preventing failures.


Subject(s)
Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Oligonucleotide Probes , Research Design/standards , Actins/genetics , Algorithms , Animals , Humans , Mice , Oligonucleotide Array Sequence Analysis/methods , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Quality Control
13.
Nucleic Acids Res ; 31(1): 94-6, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12519956

ABSTRACT

The Stanford Microarray Database (SMD; http://genome-www.stanford.edu/microarray/) serves as a microarray research database for Stanford investigators and their collaborators. In addition, SMD functions as a resource for the entire scientific community, by making freely available all of its source code and providing full public access to data published by SMD users, along with many tools to explore and analyze those data. SMD currently provides public access to data from 3500 microarrays, including data from 85 publications, and this total is increasing rapidly. In this article, we describe some of SMD's newer tools for accessing public data, assessing data quality and for data analysis.


Subject(s)
Databases, Genetic , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Animals , California , Computer Graphics , Databases, Genetic/standards , Humans , Information Storage and Retrieval , Quality Control , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...