Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Nature ; 583(7815): 265-270, 2020 07.
Article in English | MEDLINE | ID: mdl-32581361

ABSTRACT

Cancers arise through the acquisition of oncogenic mutations and grow by clonal expansion1,2. Here we reveal that most mutagenic DNA lesions are not resolved into a mutated DNA base pair within a single cell cycle. Instead, DNA lesions segregate, unrepaired, into daughter cells for multiple cell generations, resulting in the chromosome-scale phasing of subsequent mutations. We characterize this process in mutagen-induced mouse liver tumours and show that DNA replication across persisting lesions can produce multiple alternative alleles in successive cell divisions, thereby generating both multiallelic and combinatorial genetic diversity. The phasing of lesions enables accurate measurement of strand-biased repair processes, quantification of oncogenic selection and fine mapping of sister-chromatid-exchange events. Finally, we demonstrate that lesion segregation is a unifying property of exogenous mutagens, including UV light and chemotherapy agents in human cells and tumours, which has profound implications for the evolution and adaptation of cancer genomes.


Subject(s)
Chromosome Segregation/genetics , Evolution, Molecular , Genome/genetics , Neoplasms/genetics , Alleles , Animals , DNA Repair , DNA Replication , ErbB Receptors/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mutation , Neoplasms/pathology , Selection, Genetic , Signal Transduction , Sister Chromatid Exchange , Transcription, Genetic , raf Kinases/metabolism , ras Proteins/metabolism
2.
Genome Biol ; 21(1): 5, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31910870

ABSTRACT

BACKGROUND: CTCF binding contributes to the establishment of a higher-order genome structure by demarcating the boundaries of large-scale topologically associating domains (TADs). However, despite the importance and conservation of TADs, the role of CTCF binding in their evolution and stability remains elusive. RESULTS: We carry out an experimental and computational study that exploits the natural genetic variation across five closely related species to assess how CTCF binding patterns stably fixed by evolution in each species contribute to the establishment and evolutionary dynamics of TAD boundaries. We perform CTCF ChIP-seq in multiple mouse species to create genome-wide binding profiles and associate them with TAD boundaries. Our analyses reveal that CTCF binding is maintained at TAD boundaries by a balance of selective constraints and dynamic evolutionary processes. Regardless of their conservation across species, CTCF binding sites at TAD boundaries are subject to stronger sequence and functional constraints compared to other CTCF sites. TAD boundaries frequently harbor dynamically evolving clusters containing both evolutionarily old and young CTCF sites as a result of the repeated acquisition of new species-specific sites close to conserved ones. The overwhelming majority of clustered CTCF sites colocalize with cohesin and are significantly closer to gene transcription start sites than nonclustered CTCF sites, suggesting that CTCF clusters particularly contribute to cohesin stabilization and transcriptional regulation. CONCLUSIONS: Dynamic conservation of CTCF site clusters is an apparently important feature of CTCF binding evolution that is critical to the functional stability of a higher-order chromatin structure.


Subject(s)
CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Evolution, Molecular , Mice/genetics , Animals , Chromatin Immunoprecipitation Sequencing , Genome
3.
Genome Biol ; 19(1): 106, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30086769

ABSTRACT

BACKGROUND: CTCF binding to DNA helps partition the mammalian genome into discrete structural and regulatory domains. Complete removal of CTCF from mammalian cells causes catastrophic genome dysregulation, likely due to widespread collapse of 3D chromatin looping and alterations to inter- and intra-TAD interactions within the nucleus. In contrast, Ctcf hemizygous mice with lifelong reduction of CTCF expression are viable, albeit with increased cancer incidence. Here, we exploit chronic Ctcf hemizygosity to reveal its homeostatic roles in maintaining genome function and integrity. RESULTS: We find that Ctcf hemizygous cells show modest but robust changes in almost a thousand sites of genomic CTCF occupancy; these are enriched for lower affinity binding events with weaker evolutionary conservation across the mouse lineage. Furthermore, we observe dysregulation of the expression of several hundred genes, which are concentrated in cancer-related pathways, and are caused by changes in transcriptional regulation. Chromatin structure is preserved but some loop interactions are destabilized; these are often found around differentially expressed genes and their enhancers. Importantly, the transcriptional alterations identified in vitro are recapitulated in mouse tumors and also in human cancers. CONCLUSIONS: This multi-dimensional genomic and epigenomic profiling of a Ctcf hemizygous mouse model system shows that chronic depletion of CTCF dysregulates steady-state gene expression by subtly altering transcriptional regulation, changes which can also be observed in primary tumors.


Subject(s)
Breast Neoplasms/genetics , CCCTC-Binding Factor/genetics , Chromatin/chemistry , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genome , Liver Neoplasms, Experimental/genetics , Uterine Neoplasms/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CCCTC-Binding Factor/deficiency , Cell Line , Chromatin/metabolism , DNA, Neoplasm/metabolism , Enhancer Elements, Genetic , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Hemizygote , Homeostasis , Humans , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , Signal Transduction , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
4.
J Hepatol ; 69(4): 840-850, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29958939

ABSTRACT

BACKGROUND & AIMS: Carcinogen-induced mouse models of liver cancer are used extensively to study the pathogenesis of the disease and are critical for validating candidate therapeutics. These models can recapitulate molecular and histological features of human disease. However, it is not known if the genomic alterations driving these mouse tumour genomes are comparable to those found in human tumours. Herein, we provide a detailed genomic characterisation of tumours from a commonly used mouse model of hepatocellular carcinoma (HCC). METHODS: We analysed whole exome sequences of liver tumours arising in mice exposed to diethylnitrosamine (DEN). Mutational signatures were compared between liver tumours from DEN-treated and untreated mice, and human HCCs. RESULTS: DEN-initiated tumours had a high, uniform number of somatic single nucleotide variants (SNVs), with few insertions, deletions or copy number alterations, consistent with the known genotoxic action of DEN. Exposure of hepatocytes to DEN left a reproducible mutational imprint in resulting tumour exomes which we could computationally reconstruct using six known COSMIC mutational signatures. The tumours carried a high diversity of low-incidence, non-synonymous point mutations in many oncogenes and tumour suppressors, reflecting the stochastic introduction of SNVs into the hepatocyte genome by the carcinogen. We identified four recurrently mutated genes that were putative oncogenic drivers of HCC in this model. Every neoplasm carried activating hotspot mutations either in codon 61 of Hras, in codon 584 of Braf or in codon 254 of Egfr. Truncating mutations of Apc occurred in 21% of neoplasms, which were exclusively carcinomas supporting a role for deregulation of Wnt/ß-catenin signalling in cancer progression. CONCLUSIONS: Our study provides detailed insight into the mutational landscape of tumours arising in a commonly used carcinogen model of HCC, facilitating the future use of this model to better understand the human disease. LAY SUMMARY: Mouse models are widely used to study the biology of cancer and to test potential therapies. Herein, we have described the mutational landscape of tumours arising in a carcinogen-induced mouse model of liver cancer. Since cancer is a disease caused by genomic alterations, information about the patterns and types of mutations in the tumours in this mouse model should facilitate its use to study human liver cancer.


Subject(s)
Liver Neoplasms, Experimental/genetics , Mutation , Animals , DNA Copy Number Variations , Diethylnitrosamine , Disease Models, Animal , Exome , Genes, ras , Liver Neoplasms, Experimental/chemically induced , Male , Mice , Mice, Inbred C3H
5.
Genome Res ; 28(4): 448-459, 2018 04.
Article in English | MEDLINE | ID: mdl-29563166

ABSTRACT

Understanding the mechanisms driving lineage-specific evolution in both primates and rodents has been hindered by the lack of sister clades with a similar phylogenetic structure having high-quality genome assemblies. Here, we have created chromosome-level assemblies of the Mus caroli and Mus pahari genomes. Together with the Mus musculus and Rattus norvegicus genomes, this set of rodent genomes is similar in divergence times to the Hominidae (human-chimpanzee-gorilla-orangutan). By comparing the evolutionary dynamics between the Muridae and Hominidae, we identified punctate events of chromosome reshuffling that shaped the ancestral karyotype of Mus musculus and Mus caroli between 3 and 6 million yr ago, but that are absent in the Hominidae. Hominidae show between four- and sevenfold lower rates of nucleotide change and feature turnover in both neutral and functional sequences, suggesting an underlying coherence to the Muridae acceleration. Our system of matched, high-quality genome assemblies revealed how specific classes of repeats can play lineage-specific roles in related species. Recent LINE activity has remodeled protein-coding loci to a greater extent across the Muridae than the Hominidae, with functional consequences at the species level such as reproductive isolation. Furthermore, we charted a Muridae-specific retrotransposon expansion at unprecedented resolution, revealing how a single nucleotide mutation transformed a specific SINE element into an active CTCF binding site carrier specifically in Mus caroli, which resulted in thousands of novel, species-specific CTCF binding sites. Our results show that the comparison of matched phylogenetic sets of genomes will be an increasingly powerful strategy for understanding mammalian biology.


Subject(s)
Evolution, Molecular , Genome/genetics , Muridae/genetics , Phylogeny , Animals , Binding Sites , CCCTC-Binding Factor/genetics , Chromosomes/genetics , Karyotyping/methods , Long Interspersed Nucleotide Elements/genetics , Mice , Retroelements/genetics , Species Specificity
6.
Nat Commun ; 8(1): 1092, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29061983

ABSTRACT

Noncoding regulatory variants play a central role in the genetics of human diseases and in evolution. Here we measure allele-specific transcription factor binding occupancy of three liver-specific transcription factors between crosses of two inbred mouse strains to elucidate the regulatory mechanisms underlying transcription factor binding variations in mammals. Our results highlight the pre-eminence of cis-acting variants on transcription factor occupancy divergence. Transcription factor binding differences linked to cis-acting variants generally exhibit additive inheritance, while those linked to trans-acting variants are most often dominantly inherited. Cis-acting variants lead to local coordination of transcription factor occupancies that decay with distance; distal coordination is also observed and may be modulated by long-range chromatin contacts. Our results reveal the regulatory mechanisms that interplay to drive transcription factor occupancy, chromatin state, and gene expression in complex mammalian cell states.


Subject(s)
Chromatin/metabolism , Transcription Factors/metabolism , Alleles , Animals , Chromatin/genetics , Evolution, Molecular , Gene Expression Regulation, Fungal/genetics , Gene Expression Regulation, Fungal/physiology , Humans , Mice , Protein Binding/genetics , Protein Binding/physiology , Transcription Factors/genetics
7.
J Exp Med ; 214(3): 579-596, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28232471

ABSTRACT

Pancreatic stellate cells (PSCs) differentiate into cancer-associated fibroblasts (CAFs) that produce desmoplastic stroma, thereby modulating disease progression and therapeutic response in pancreatic ductal adenocarcinoma (PDA). However, it is unknown whether CAFs uniformly carry out these tasks or if subtypes of CAFs with distinct phenotypes in PDA exist. We identified a CAF subpopulation with elevated expression of α-smooth muscle actin (αSMA) located immediately adjacent to neoplastic cells in mouse and human PDA tissue. We recapitulated this finding in co-cultures of murine PSCs and PDA organoids, and demonstrated that organoid-activated CAFs produced desmoplastic stroma. The co-cultures showed cooperative interactions and revealed another distinct subpopulation of CAFs, located more distantly from neoplastic cells, which lacked elevated αSMA expression and instead secreted IL6 and additional inflammatory mediators. These findings were corroborated in mouse and human PDA tissue, providing direct evidence for CAF heterogeneity in PDA tumor biology with implications for disease etiology and therapeutic development.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Fibroblasts/physiology , Myofibroblasts/physiology , Pancreatic Neoplasms/pathology , Actins/analysis , Animals , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Cells, Cultured , Cytokines/biosynthesis , Humans , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , STAT3 Transcription Factor/metabolism
8.
J Pathol ; 239(3): 286-96, 2016 07.
Article in English | MEDLINE | ID: mdl-27061193

ABSTRACT

Stromal targeting for pancreatic ductal adenocarcinoma (PDAC) is rapidly becoming an attractive option, due to the lack of efficacy of standard chemotherapy and increased knowledge about PDAC stroma. We postulated that the addition of stromal therapy may enhance the anti-tumour efficacy of chemotherapy. Gemcitabine and all-trans retinoic acid (ATRA) were combined in a clinically applicable regimen, to target cancer cells and pancreatic stellate cells (PSCs) respectively, in 3D organotypic culture models and genetically engineered mice (LSL-Kras(G12D) (/+) ;LSL-Trp53(R172H) (/+) ;Pdx-1-Cre: KPC mice) representing the spectrum of PDAC. In two distinct sets of organotypic models as well as KPC mice, we demonstrate a reduction in cancer cell proliferation and invasion together with enhanced cancer cell apoptosis when ATRA is combined with gemcitabine, compared to vehicle or either agent alone. Simultaneously, PSC activity (as measured by deposition of extracellular matrix proteins such as collagen and fibronectin) and PSC invasive ability were both diminished in response to combination therapy. These effects were mediated through a range of signalling cascades (Wnt, hedgehog, retinoid, and FGF) in cancer as well as stellate cells, affecting epithelial cellular functions such as epithelial-mesenchymal transition, cellular polarity, and lumen formation. At the tissue level, this resulted in enhanced tumour necrosis, increased vascularity, and diminished hypoxia. Consequently, there was an overall reduction in tumour size. The enhanced effect of stromal co-targeting (ATRA) alongside chemotherapy (gemcitabine) appears to be mediated by dampening multiple signalling cascades in the tumour-stroma cross-talk, rather than ablating stroma or targeting a single pathway. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Adenocarcinoma/therapy , Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/therapy , Signal Transduction/drug effects , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/therapeutic use , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Humans , Mice , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/pathology , Gemcitabine
9.
Plast Reconstr Surg Glob Open ; 2(11): e249, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25506532

ABSTRACT

BACKGROUND: Preoperative signs and symptoms of patients with Poly Implant Prothese (PIP) implants could be predictive of device failure. Based on clinical observation and intraoperative findings 4 hypotheses were raised: (1) Preoperative clinical signs including acquired asymmetry, breast enlargement, fullness of the lower pole, decreased mound projection, and change in breast consistency could be indicative of implant rupture. (2) Device failure correlates with a low preoperative Baker grade of capsule. (3) Brown-stained implants are more prone to implant failure. (4) The brown gel could be indicative of iodine ingression through a substandard elastomer shell. METHODS: Preoperative clinical signs were compared with intraoperative findings for 27 patients undergoing PIP implant explantation. RESULTS: Acquired asymmetry (P = 0.0003), breast enlargement (P = 0.0002), fuller lower pole (P < 0.0001), and loss of lateral projection (P < 0.0001) were all significantly predictive of device failure. Capsule Baker grade was lower preoperatively for ruptured implants. The lack of palpable and visible preoperative capsular contracture could be secondary to the elastic nature of the capsular tissue found. Brown implants failed significantly more often than white implants. Analysis of brown gel revealed the presence of iodine, suggesting povidone iodine ingression at implantation. CONCLUSIONS: Preoperative signs can be predictive of PIP implant failure. Brown-stained implants are more prone to rupture. The presence of iodine in the gel suggests unacceptable permeability of the shell early in the implant's life span. A noninvasive screening test to detect brown implants in situ could help identify implants at risk of failure in those who elect to keep their implants.

10.
Proc Natl Acad Sci U S A ; 110(50): 20212-7, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277834

ABSTRACT

An autochthonous model of pancreatic ductal adenocarcinoma (PDA) permitted the analysis of why immunotherapy is ineffective in this human disease. Despite finding that PDA-bearing mice had cancer cell-specific CD8(+) T cells, the mice, like human patients with PDA, did not respond to two immunological checkpoint antagonists that promote the function of T cells: anti-cytotoxic T-lymphocyte-associated protein 4 (α-CTLA-4) and α-programmed cell death 1 ligand 1 (α-PD-L1). Immune control of PDA growth was achieved, however, by depleting carcinoma-associated fibroblasts (CAFs) that express fibroblast activation protein (FAP). The depletion of the FAP(+) stromal cell also uncovered the antitumor effects of α-CTLA-4 and α-PD-L1, indicating that its immune suppressive activity accounts for the failure of these T-cell checkpoint antagonists. Three findings suggested that chemokine (C-X-C motif) ligand 12 (CXCL12) explained the overriding immunosuppression by the FAP(+) cell: T cells were absent from regions of the tumor containing cancer cells, cancer cells were coated with the chemokine, CXCL12, and the FAP(+) CAF was the principal source of CXCL12 in the tumor. Administering AMD3100, a CXCL12 receptor chemokine (C-X-C motif) receptor 4 inhibitor, induced rapid T-cell accumulation among cancer cells and acted synergistically with α-PD-L1 to greatly diminish cancer cells, which were identified by their loss of heterozygosity of Trp53 gene. The residual tumor was composed only of premalignant epithelial cells and inflammatory cells. Thus, a single protein, CXCL12, from a single stromal cell type, the FAP(+) CAF, may direct tumor immune evasion in a model of human PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Chemokine CXCL12/metabolism , Gelatinases/metabolism , Immunotherapy/methods , Membrane Proteins/metabolism , Pancreatic Neoplasms/therapy , Serine Endopeptidases/metabolism , Tumor Escape/genetics , Analysis of Variance , Animals , Base Sequence , Benzylamines , Carcinoma, Pancreatic Ductal/immunology , Cyclams , Endopeptidases , Enzyme-Linked Immunospot Assay , Fibroblasts/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Heterocyclic Compounds , Immunohistochemistry , Mice , Molecular Sequence Data , Pancreatic Neoplasms/immunology , Sequence Analysis, RNA
11.
EMBO J ; 32(24): 3114-5, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24270571

ABSTRACT

Cohesin is a conserved protein complex indispensible for proper cell division, because it secures sister-chromatid cohesion following DNA replication until segregation is required at the onset of anaphase. Recent studies have revealed functions beyond this, showing that cohesin binds to interphase chromatin regulating gene expression at select loci via long-range chromosomal interactions. In this issue of The EMBO Journal, Sofueva et al (2013) use a combination of chromatin conformation capture methods, classical FISH imaging, and loss-of-function studies to elegantly demonstrate how cohesin controls the 3D architectural organization of the genome.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/chemistry , Chromosomes/metabolism , Animals , Cohesins
12.
J Exp Med ; 210(6): 1137-51, 2013 Jun 03.
Article in English | MEDLINE | ID: mdl-23712428

ABSTRACT

Fibroblast activation protein-α (FAP) identifies stromal cells of mesenchymal origin in human cancers and chronic inflammatory lesions. In mouse models of cancer, they have been shown to be immune suppressive, but studies of their occurrence and function in normal tissues have been limited. With a transgenic mouse line permitting the bioluminescent imaging of FAP(+) cells, we find that they reside in most tissues of the adult mouse. FAP(+) cells from three sites, skeletal muscle, adipose tissue, and pancreas, have highly similar transcriptomes, suggesting a shared lineage. FAP(+) cells of skeletal muscle are the major local source of follistatin, and in bone marrow they express Cxcl12 and KitL. Experimental ablation of these cells causes loss of muscle mass and a reduction of B-lymphopoiesis and erythropoiesis, revealing their essential functions in maintaining normal muscle mass and hematopoiesis, respectively. Remarkably, these cells are altered at these sites in transplantable and spontaneous mouse models of cancer-induced cachexia and anemia. Thus, the FAP(+) stromal cell may have roles in two adverse consequences of cancer: their acquisition by tumors may cause failure of immunosurveillance, and their alteration in normal tissues contributes to the paraneoplastic syndromes of cachexia and anemia.


Subject(s)
Anemia/metabolism , Bone Marrow/metabolism , Cachexia/metabolism , Gelatinases/genetics , Gelatinases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle, Skeletal/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Stromal Cells/metabolism , Adipose Tissue/metabolism , Anemia/genetics , Anemia/pathology , Animals , Cachexia/genetics , Cachexia/pathology , Cell Lineage/genetics , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Endopeptidases , Erythropoiesis/genetics , Follistatin/genetics , Follistatin/metabolism , Hematopoiesis/genetics , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Muscle, Skeletal/cytology , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Pancreas/metabolism , Stromal Cells/cytology , Transcriptome/genetics
13.
Gut ; 62(1): 112-20, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22466618

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDA) is characterised by stromal desmoplasia and vascular dysfunction, which critically impair drug delivery. This study examines the role of an abundant extracellular matrix component, the megadalton glycosaminoglycan hyaluronan (HA), as a novel therapeutic target in PDA. METHODS: Using a genetically engineered mouse model of PDA, the authors enzymatically depleted HA by a clinically formulated PEGylated human recombinant PH20 hyaluronidase (PEGPH20) and examined tumour perfusion, vascular permeability and drug delivery. The preclinical utility of PEGPH20 in combination with gemcitabine was assessed by short-term and survival studies. RESULTS: PEGPH20 rapidly and sustainably depleted HA, inducing the re-expansion of PDA blood vessels and increasing the intratumoral delivery of two chemotherapeutic agents, doxorubicin and gemcitabine. Moreover, PEGPH20 triggered fenestrations and interendothelial junctional gaps in PDA tumour endothelia and promoted a tumour-specific increase in macromolecular permeability. Finally, combination therapy with PEGPH20 and gemcitabine led to inhibition of PDA tumour growth and prolonged survival over gemcitabine monotherapy, suggesting immediate clinical utility. CONCLUSIONS: The authors demonstrate that HA impedes the intratumoral vasculature in PDA and propose that its enzymatic depletion be explored as a means to improve drug delivery and response in patients with pancreatic cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/physiology , Carcinoma, Pancreatic Ductal/drug therapy , Drug Delivery Systems , Drug Resistance, Neoplasm/physiology , Hyaluronic Acid/physiology , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/physiopathology , Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/pharmacology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm/drug effects , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/pharmacology , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Mice, Transgenic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/physiopathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Tissue Array Analysis , Treatment Outcome , Gemcitabine
14.
Clin Cancer Res ; 18(16): 4266-76, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22896693

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a common and lethal malignancy resulting in more than 250,000 deaths per year worldwide. Despite extensive efforts, cytotoxic and targeted therapies have provided only limited efficacy for patients with PDA to date. One contributing factor to the failure of systemic therapies may be the abundant tumor stromal content that is the characteristic of PDA. The PDA stroma, aptly termed the tumor microenvironment, occupies the majority of the tumor mass, and consists of a dynamic assortment of extracellular matrix components and nonneoplastic cells including fibroblastic, vascular, and immune cells. Recent work has revealed that the PDA stroma supports tumor growth and promotes metastasis and simultaneously serves as a physical barrier to drug delivery. Accordingly, methods that alter stromal composition or function, for instance interference with the vasculature via Notch/Hedgehog pathway inhibition or relief of vascular compression by hyaluronidase, are under active investigation. Here, we will review our current understanding of the PDA tumor microenvironment, and highlight opportunities for further exploration that may benefit patients.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Extracellular Matrix , Fibroblasts , Humans , Hypoxia , Pancreatic Neoplasms/drug therapy , Pancreatic Stellate Cells , Stromal Cells , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
15.
Gastroenterology ; 141(4): 1486-97, 1497.e1-14, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21704588

ABSTRACT

BACKGROUND & AIMS: Patients with pancreatic ductal adenocarcinoma are deficient in vitamin A, resulting in activation of pancreatic stellate cells (PSCs). We investigated whether restoration of retinol to PSCs restores their quiescence and affects adjacent cancer cells. METHODS: PSCs and cancer cell lines (AsPc1 and Capan1) were exposed to doses and isoforms of retinoic acid (RA) in 2-dimensional and 3-dimensional culture conditions (physiomimetic organotypic culture). The effects of all-trans retinoic acid (ATRA) were studied in LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre mice, a model of human pancreatic ductal adenocarcinoma. RESULTS: After incubation with ATRA, PSCs were quiescent and had altered expression of genes that regulate proliferation, morphology, and motility; genes that encode cytoskeletal proteins and cytokines; and genes that control other functions, irrespective of culture conditions or dosage. In the organotypic model, and in mice, ATRA induced quiescence of PSCs and thereby reduced cancer cell proliferation and translocation of ß-catenin to the nucleus, increased cancer cell apoptosis, and altered tumor morphology. ATRA reduced the motility of PSCs, so these cells created a "wall" at the junction between the tumor and the matrix that prevented cancer cell invasion. Restoring secreted frizzled-related protein 4 (sFRP4) secretion to quiescent PSCs reduced Wnt-ß-catenin signaling in cancer cells and their invasive ability. Human primary and metastatic pancreatic tumor tissues stained strongly for cancer cell nuclear ß-catenin but had low levels of sFRP4 (in cancer cells and PSCs). CONCLUSIONS: RA induces quiescence and reduces motility of PSCs, leading to reduced proliferation and increased apoptosis of surrounding pancreatic cancer cells. RA isoforms might be developed as therapeutic reagents for pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Cellular Senescence/drug effects , Pancreatic Neoplasms/drug therapy , Pancreatic Stellate Cells/drug effects , Paracrine Communication/drug effects , Signal Transduction/drug effects , Tretinoin/pharmacology , Wnt Proteins/metabolism , beta Catenin/metabolism , Alitretinoin , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isotretinoin/pharmacology , Mice , Mice, Mutant Strains , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Proto-Oncogene Proteins/metabolism , RNA Interference , Time Factors , Transcription, Genetic/drug effects
16.
Gut ; 60(6): 861-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20966025

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is an almost uniformly lethal disease. One explanation for the devastating prognosis is the failure of many chemotherapies, including the current standard of care therapy gemcitabine. Although our knowledge of the molecular events underlying multistep carcinogenesis in PDA has steadily increased, translation into more effective therapeutic approaches has been inefficient over the last several decades. Evidence for this innate resistance to systemic therapies was recently provided in an accurate mouse model of PDA by the demonstration that chemotherapies are poorly delivered to PDA tissues because of a deficient vasculature. This vascular deficiency correlated with the presence of a dense stromal matrix that is a prominent histological hallmark of PDA tumours. Therapeutic targeting of stromal cells decreased the stroma from pancreatic tumours, resulting in increased intratumoral perfusion and therapeutic delivery of gemcitabine. Stromal cells contained within the PDA tumour microenvironment therefore represent an additional constituent to neoplastic cells that should be critically evaluated for optimal therapeutic development in preclinical models and early clinical trials.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Stromal Cells/pathology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Genetic Engineering/methods , Humans , Mice , Mice, Mutant Strains , Pancreatic Neoplasms/pathology , Stromal Cells/drug effects , Tumor Microenvironment/physiology
17.
Nature ; 465(7297): 492-6, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20505730

ABSTRACT

CD95 (also called Fas and APO-1) is a prototypical death receptor that regulates tissue homeostasis mainly in the immune system through the induction of apoptosis. During cancer progression CD95 is frequently downregulated or cells are rendered apoptosis resistant, raising the possibility that loss of CD95 is part of a mechanism for tumour evasion. However, complete loss of CD95 is rarely seen in human cancers and many cancer cells express large quantities of CD95 and are highly sensitive to CD95-mediated apoptosis in vitro. Furthermore, cancer patients frequently have elevated levels of the physiological ligand for CD95, CD95L. These data raise the possibility that CD95 could actually promote the growth of tumours through its non-apoptotic activities. Here we show that cancer cells in general, regardless of their CD95 apoptosis sensitivity, depend on constitutive activity of CD95, stimulated by cancer-produced CD95L, for optimal growth. Consistently, loss of CD95 in mouse models of ovarian cancer and liver cancer reduces cancer incidence as well as the size of the tumours. The tumorigenic activity of CD95 is mediated by a pathway involving JNK and Jun. These results demonstrate that CD95 has a growth-promoting role during tumorigenesis and indicate that efforts to inhibit its activity rather than to enhance it should be considered during cancer therapy.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , fas Receptor/metabolism , Animals , Apoptosis , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Cell Line, Tumor , Cell Proliferation , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/immunology , Fas Ligand Protein/metabolism , Female , Gene Expression Regulation, Neoplastic , Hepatocytes/enzymology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , fas Receptor/deficiency , fas Receptor/genetics
18.
Science ; 324(5933): 1457-61, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19460966

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is among the most lethal human cancers in part because it is insensitive to many chemotherapeutic drugs. Studying a mouse model of PDA that is refractory to the clinically used drug gemcitabine, we found that the tumors in this model were poorly perfused and poorly vascularized, properties that are shared with human PDA. We tested whether the delivery and efficacy of gemcitabine in the mice could be improved by coadministration of IPI-926, a drug that depletes tumor-associated stromal tissue by inhibition of the Hedgehog cellular signaling pathway. The combination therapy produced a transient increase in intratumoral vascular density and intratumoral concentration of gemcitabine, leading to transient stabilization of disease. Thus, inefficient drug delivery may be an important contributor to chemoresistance in pancreatic cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Hedgehog Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Veratrum Alkaloids/administration & dosage , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/metabolism , Deoxycytidine/therapeutic use , Disease Models, Animal , Drug Resistance, Neoplasm , Hedgehog Proteins/antagonists & inhibitors , Humans , Kruppel-Like Transcription Factors/metabolism , Mice , Neoplasm Transplantation , Neovascularization, Pathologic , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Smoothened Receptor , Stromal Cells/drug effects , Stromal Cells/pathology , Veratrum Alkaloids/pharmacokinetics , Veratrum Alkaloids/therapeutic use , Zinc Finger Protein GLI1 , Gemcitabine
19.
Methods Enzymol ; 442: 83-100, 2008.
Article in English | MEDLINE | ID: mdl-18662565

ABSTRACT

Major work elucidating the structure and function of the CD95 death inducing signaling complex (DISC) was carried in the late 1990s. Since then the DISC has become a paradigm for multiprotein signaling complexes in the apoptosis literature. We analyzed the earliest events of DISC formation with a set of different techniques and found a surprising new characteristic of the CD95 DISC. Data revealed that CD95 is palmitoylated on cysteine 199. This lipid modification enhances receptor aggregation to the high molecular weight DISC.


Subject(s)
Apoptosis/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , fas Receptor/physiology , Animals , Blotting, Western , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Immunoprecipitation , Lipoylation , Molecular Weight , Ultracentrifugation , fas Receptor/chemistry , fas Receptor/metabolism
20.
Cancer Res ; 68(8): 2587-91, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413726

ABSTRACT

MicroRNAs (miRNA) are small RNA molecules of approximately 20 to 22 nucleotides that reduce expression of proteins through mRNA degradation and/or translational silencing. Each known miRNA has a large number of predicted targets. Members of the let-7/miR-98 family of miRNAs are up-regulated at the end of embryonic development. Let-7 is often down-regulated early during cancer development, suggesting that let-7-regulated oncofetal genes (LOG) may become reexpressed in cancer cells. Using comparative bioinformatics, we have identified 12 conserved LOGs that include HMGA2 and IMP-1/CRD-BP. IMP-1 has growth-promoting activities through stabilization of c-myc mRNA. We experimentally confirmed that IMP-1 is a direct let-7 target that promotes cell growth and motility of tumor cells, and we confirmed by proteomics analysis that IMP-1 and HMGA2 are major miRNA targets. Our data suggest that a substantial part of the growth inhibitory activities of let-7 comes from suppressing the expression of IMP-1. LOGs could be novel therapeutic targets and potential biomarkers for cancer treatment.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Adenocarcinoma , Animals , Antigens, Neoplasm/genetics , Cell Division , Cell Line, Tumor , Cell Movement , DNA Primers , Gene Silencing , Humans , Lung Neoplasms , Mice , Polymerase Chain Reaction , RNA, Messenger/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...