Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
Toxics ; 12(5)2024 Apr 29.
Article En | MEDLINE | ID: mdl-38787102

Artisanal and small-scale gold mining (ASGM) plays a crucial role in global gold production. However, the adoption of poor mining practices or the use of mercury (Hg) in gold recovery processes has generated serious environmental contamination events. The focus of this study is assessing the concentration of Hg in surface waters within the coastal region of Ecuador. The results are used to conduct a human health risk assessment applying deterministic and probabilistic methods, specifically targeting groups vulnerable to exposure in affected mining environments. Between April and June 2022, 54 water samples were collected from rivers and streams adjacent to mining areas to determine Hg levels. In the health risk assessment, exposure routes through water ingestion and dermal contact were considered for both adults and children, following the model structures outlined by the U.S. Environmental Protection Agency. The results indicate elevated Hg concentrations in two of the five provinces studied, El Oro and Esmeraldas, where at least 88% and 75% of the samples, respectively, exceeded the maximum permissible limit (MPL) set by Ecuadorian regulations for the preservation of aquatic life. Furthermore, in El Oro province, 28% of the samples exceeded the MPL established for drinking water quality. The high concentrations of Hg could be related to illegal mining activity that uses Hg for gold recovery. Regarding the human health risk assessment, risk values above the safe exposure limit were estimated. Children were identified as the most vulnerable receptor. Therefore, there is an urgent need to establish effective regulations that guarantee the protection of river users in potentially contaminated areas. Finally, it is important to continue investigating the contamination caused by human practices in the coastal region.

2.
Nat Biotechnol ; 42(3): 424-436, 2024 Mar.
Article En | MEDLINE | ID: mdl-37169967

Genetically engineered mouse models only capture a small fraction of the genetic lesions that drive human cancer. Current CRISPR-Cas9 models can expand this fraction but are limited by their reliance on error-prone DNA repair. Here we develop a system for in vivo prime editing by encoding a Cre-inducible prime editor in the mouse germline. This model allows rapid, precise engineering of a wide range of mutations in cell lines and organoids derived from primary tissues, including a clinically relevant Kras mutation associated with drug resistance and Trp53 hotspot mutations commonly observed in pancreatic cancer. With this system, we demonstrate somatic prime editing in vivo using lipid nanoparticles, and we model lung and pancreatic cancer through viral delivery of prime editing guide RNAs or orthotopic transplantation of prime-edited organoids. We believe that this approach will accelerate functional studies of cancer-associated mutations and complex genetic combinations that are challenging to construct with traditional models.


Pancreatic Neoplasms , RNA, Guide, CRISPR-Cas Systems , Mice , Humans , Animals , Mice, Transgenic , Mutation/genetics , Pancreatic Neoplasms/genetics , Cell Line , Gene Editing , CRISPR-Cas Systems/genetics
3.
Nat Genet ; 55(10): 1686-1695, 2023 10.
Article En | MEDLINE | ID: mdl-37709863

DNA mismatch repair deficiency (MMRd) is associated with a high tumor mutational burden (TMB) and sensitivity to immune checkpoint blockade (ICB) therapy. Nevertheless, most MMRd tumors do not durably respond to ICB and critical questions remain about immunosurveillance and TMB in these tumors. In the present study, we developed autochthonous mouse models of MMRd lung and colon cancer. Surprisingly, these models did not display increased T cell infiltration or ICB response, which we showed to be the result of substantial intratumor heterogeneity of mutations. Furthermore, we found that immunosurveillance shapes the clonal architecture but not the overall burden of neoantigens, and T cell responses against subclonal neoantigens are blunted. Finally, we showed that clonal, but not subclonal, neoantigen burden predicts ICB response in clinical trials of MMRd gastric and colorectal cancer. These results provide important context for understanding immune evasion in cancers with a high TMB and have major implications for therapies aimed at increasing TMB.


Brain Neoplasms , Colorectal Neoplasms , Neoplastic Syndromes, Hereditary , Animals , Mice , Colorectal Neoplasms/genetics , Antigens, Neoplasm/genetics , Mutation , DNA Mismatch Repair/genetics , Biomarkers, Tumor/genetics
4.
Cancer Cell ; 41(5): 871-886.e10, 2023 05 08.
Article En | MEDLINE | ID: mdl-37059105

Lymphocytes are key for immune surveillance of tumors, but our understanding of the spatial organization and physical interactions that facilitate lymphocyte anti-cancer functions is limited. We used multiplexed imaging, quantitative spatial analysis, and machine learning to create high-definition maps of lung tumors from a Kras/Trp53-mutant mouse model and human resections. Networks of interacting lymphocytes ("lymphonets") emerged as a distinctive feature of the anti-cancer immune response. Lymphonets nucleated from small T cell clusters and incorporated B cells with increasing size. CXCR3-mediated trafficking modulated lymphonet size and number, but T cell antigen expression directed intratumoral localization. Lymphonets preferentially harbored TCF1+ PD-1+ progenitor CD8+ T cells involved in responses to immune checkpoint blockade (ICB) therapy. Upon treatment of mice with ICB or an antigen-targeted vaccine, lymphonets retained progenitor and gained cytotoxic CD8+ T cell populations, likely via progenitor differentiation. These data show that lymphonets create a spatial environment supportive of CD8+ T cell anti-tumor responses.


Adenocarcinoma of Lung , Lung Neoplasms , Humans , Animals , Mice , CD8-Positive T-Lymphocytes , Immunotherapy/methods , Lung Neoplasms/pathology , Adenocarcinoma of Lung/genetics , Immunity
5.
Nat Commun ; 13(1): 5745, 2022 10 03.
Article En | MEDLINE | ID: mdl-36192379

Diverse processes in cancer are mediated by enzymes, which most proximally exert their function through their activity. High-fidelity methods to profile enzyme activity are therefore critical to understanding and targeting the pathological roles of enzymes in cancer. Here, we present an integrated set of methods for measuring specific protease activities across scales, and deploy these methods to study treatment response in an autochthonous model of Alk-mutant lung cancer. We leverage multiplexed nanosensors and machine learning to analyze in vivo protease activity dynamics in lung cancer, identifying significant dysregulation that includes enhanced cleavage of a peptide, S1, which rapidly returns to healthy levels with targeted therapy. Through direct on-tissue localization of protease activity, we pinpoint S1 cleavage to the tumor vasculature. To link protease activity to cellular function, we design a high-throughput method to isolate and characterize proteolytically active cells, uncovering a pro-angiogenic phenotype in S1-cleaving cells. These methods provide a framework for functional, multiscale characterization of protease dysregulation in cancer.


Lung Neoplasms , Peptide Hydrolases , Endopeptidases , Humans , Lung Neoplasms/genetics , Peptide Hydrolases/metabolism , Proteolysis , Receptor Protein-Tyrosine Kinases
6.
Genes Dev ; 36(15-16): 936-949, 2022 08 01.
Article En | MEDLINE | ID: mdl-36175034

Lung cancer is the leading cause of cancer-related death worldwide. Lung adenocarcinoma (LUAD), the most common histological subtype, accounts for 40% of all cases. While existing genetically engineered mouse models (GEMMs) recapitulate the histological progression and transcriptional evolution of human LUAD, they are time-consuming and technically demanding. In contrast, cell line transplant models are fast and flexible, but these models fail to capture the full spectrum of disease progression. Organoid technologies provide a means to create next-generation cancer models that integrate the most advantageous features of autochthonous and transplant-based systems. However, robust and faithful LUAD organoid platforms are currently lacking. Here, we describe optimized conditions to continuously expand murine alveolar type 2 (AT2) cells, a prominent cell of origin for LUAD, in organoid culture. These organoids display canonical features of AT2 cells, including marker gene expression, the presence of lamellar bodies, and an ability to differentiate into the AT1 lineage. We used this system to develop flexible and versatile immunocompetent organoid-based models of KRAS, BRAF, and ALK mutant LUAD. Notably, organoid-based tumors display extensive burden and complete penetrance and are histopathologically indistinguishable from their autochthonous counterparts. Altogether, this organoid platform is a powerful, versatile new model system to study LUAD.


Adenocarcinoma of Lung , Lung Neoplasms , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Humans , Lung Neoplasms/metabolism , Mice , Organoids , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
7.
Nature ; 607(7917): 149-155, 2022 07.
Article En | MEDLINE | ID: mdl-35705813

Immunosurveillance of cancer requires the presentation of peptide antigens on major histocompatibility complex class I (MHC-I) molecules1-5. Current approaches to profiling of MHC-I-associated peptides, collectively known as the immunopeptidome, are limited to in vitro investigation or bulk tumour lysates, which limits our understanding of cancer-specific patterns of antigen presentation in vivo6. To overcome these limitations, we engineered an inducible affinity tag into the mouse MHC-I gene (H2-K1) and targeted this allele to the KrasLSL-G12D/+Trp53fl/fl mouse model (KP/KbStrep)7. This approach enabled us to precisely isolate MHC-I peptides from autochthonous pancreatic ductal adenocarcinoma and from lung adenocarcinoma (LUAD) in vivo. In addition, we profiled the LUAD immunopeptidome from the alveolar type 2 cell of origin up to late-stage disease. Differential peptide presentation in LUAD was not predictable by mRNA expression or translation efficiency and is probably driven by post-translational mechanisms. Vaccination with peptides presented by LUAD in vivo induced CD8+ T cell responses in naive mice and tumour-bearing mice. Many peptides specific to LUAD, including immunogenic peptides, exhibited minimal expression of the cognate mRNA, which prompts the reconsideration of antigen prediction pipelines that triage peptides according to transcript abundance8. Beyond cancer, the KbStrep allele is compatible with other Cre-driver lines to explore antigen presentation in vivo in the pursuit of understanding basic immunology, infectious disease and autoimmunity.


Antigens, Neoplasm , Peptides , Proteomics , Alveolar Epithelial Cells/immunology , Animals , Antigen Presentation , Antigens, Neoplasm/analysis , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/chemistry , Carcinoma, Pancreatic Ductal/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Lung Neoplasms/chemistry , Lung Neoplasms/immunology , Mice , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/immunology , Peptides/analysis , Peptides/chemistry , Peptides/immunology , RNA, Messenger
8.
Cell ; 185(11): 1905-1923.e25, 2022 05 26.
Article En | MEDLINE | ID: mdl-35523183

Tumor evolution is driven by the progressive acquisition of genetic and epigenetic alterations that enable uncontrolled growth and expansion to neighboring and distal tissues. The study of phylogenetic relationships between cancer cells provides key insights into these processes. Here, we introduced an evolving lineage-tracing system with a single-cell RNA-seq readout into a mouse model of Kras;Trp53(KP)-driven lung adenocarcinoma and tracked tumor evolution from single-transformed cells to metastatic tumors at unprecedented resolution. We found that the loss of the initial, stable alveolar-type2-like state was accompanied by a transient increase in plasticity. This was followed by the adoption of distinct transcriptional programs that enable rapid expansion and, ultimately, clonal sweep of stable subclones capable of metastasizing. Finally, tumors develop through stereotypical evolutionary trajectories, and perturbing additional tumor suppressors accelerates progression by creating novel trajectories. Our study elucidates the hierarchical nature of tumor evolution and, more broadly, enables in-depth studies of tumor progression.


Neoplasms , Animals , Genes, ras , Mice , Neoplasms/genetics , Phylogeny , Exome Sequencing
9.
Cancer Cell ; 38(2): 212-228.e13, 2020 08 10.
Article En | MEDLINE | ID: mdl-32707078

Regulatory networks that maintain functional, differentiated cell states are often dysregulated in tumor development. Here, we use single-cell epigenomics to profile chromatin state transitions in a mouse model of lung adenocarcinoma (LUAD). We identify an epigenomic continuum representing loss of cellular identity and progression toward a metastatic state. We define co-accessible regulatory programs and infer key activating and repressive chromatin regulators of these cell states. Among these co-accessibility programs, we identify a pre-metastatic transition, characterized by activation of RUNX transcription factors, which mediates extracellular matrix remodeling to promote metastasis and is predictive of survival across human LUAD patients. Together, these results demonstrate the power of single-cell epigenomics to identify regulatory programs to uncover mechanisms and key biomarkers of tumor progression.


Adenocarcinoma/genetics , Disease Models, Animal , Epigenomics/methods , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Disease Progression , Gene Expression Profiling/methods , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/genetics , Single-Cell Analysis/methods
10.
Nat Cancer ; 1(6): 589-602, 2020 06.
Article En | MEDLINE | ID: mdl-34414377

Approximately 20-30% of human lung adenocarcinomas (LUAD) harbor loss-of-function (LOF) mutations in Kelch-like ECH Associated-Protein 1 (KEAP1), which lead to hyperactivation of the nuclear factor, erythroid 2-like 2 (NRF2) antioxidant pathway and correlate with poor prognosis1-3. We previously showed that Keap1 mutation accelerates KRAS-driven LUAD and produces a marked dependency on glutaminolysis4. To extend the investigation of genetic dependencies in the context of Keap1 mutation, we performed a druggable genome CRISPR-Cas9 screen in Keap1-mutant cells. This analysis uncovered a profound Keap1 mutant-specific dependency on solute carrier family 33 member 1 (Slc33a1), an endomembrane-associated protein with roles in autophagy regulation5, as well as a series of functionally-related genes implicated in the unfolded protein response. Targeted genetic and biochemical experiments using mouse and human Keap1-mutant tumor lines, as well as preclinical genetically-engineered mouse models (GEMMs) of LUAD, validate Slc33a1 as a robust Keap1-mutant-specific dependency. Furthermore, unbiased genome-wide CRISPR screening identified additional genes related to Slc33a1 dependency. Overall, our study provides a strong rationale for stratification of patients harboring KEAP1-mutant or NRF2-hyperactivated tumors as likely responders to targeted SLC33A1 inhibition and underscores the value of integrating functional genetic approaches with GEMMs to identify and validate genotype-specific therapeutic targets.


Adenocarcinoma of Lung , Kelch-Like ECH-Associated Protein 1 , Lung Neoplasms , Membrane Transport Proteins , Adenocarcinoma of Lung/genetics , Animals , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Lung Neoplasms/genetics , Membrane Transport Proteins/genetics , Mice , Mutation , NF-E2-Related Factor 2/genetics
12.
Cornea ; 39(5): 535-539, 2020 May.
Article En | MEDLINE | ID: mdl-31651431

PURPOSE: To present the results of corneal wedge resection in postkeratoplasty astigmatism, performed by the same surgeon using the same nomogram over a 25-year period. METHODS: This is a retrospective observational study. The sample was obtained from the medical records of all patients who underwent penetrating or deep lamellar keratoplasty, performed by a single surgeon from 1993 to 2018. All surgeries were performed using a diamond knife, on the flat meridian, involving the keratoplasty scar and closed with five 10-0 nylon sutures. RESULTS: A total of 39 eyes were included. The keratometry measured cylinder improved from 7.99 ± 0.25 to 2.5 ± 0.3 D at 12 months and remained stable thereafter (a mean follow-up of 76.3 months). Best spectacle corrected visual acuity increased from 0.35 ± 0.01 to 0.57 ± 0.02 at 12 months and remained stable thereafter. There was a coupling ratio of 0.08 ± 0.03 D at 12 months. There were no corneal graft rejections or loss of best spectacle corrected visual acuity on this series. CONCLUSIONS: Corneal wedge resection is a valuable resource for the management of high postkeratoplasty astigmatism. It is a safe and reproducible procedure, with stable results at 12 months and thereafter.


Astigmatism/surgery , Cornea/surgery , Keratoplasty, Penetrating/adverse effects , Postoperative Complications/surgery , Refraction, Ocular/physiology , Visual Acuity , Adult , Aged , Astigmatism/diagnosis , Astigmatism/etiology , Cornea/pathology , Corneal Topography , Female , Follow-Up Studies , Humans , Male , Middle Aged , Postoperative Complications/diagnosis , Postoperative Complications/etiology , Reoperation , Retrospective Studies
13.
Nature ; 571(7765): 398-402, 2019 07.
Article En | MEDLINE | ID: mdl-31292548

A decline in stem cell function impairs tissue regeneration during ageing, but the role of the stem-cell-supporting niche in ageing is not well understood. The small intestine is maintained by actively cycling intestinal stem cells that are regulated by the Paneth cell niche1,2. Here we show that the regenerative potential of human and mouse intestinal epithelium diminishes with age owing to defects in both stem cells and their niche. The functional decline was caused by a decrease in stemness-maintaining Wnt signalling due to production of Notum, an extracellular Wnt inhibitor, in aged Paneth cells. Mechanistically, high activity of mammalian target of rapamycin complex 1 (mTORC1) in aged Paneth cells inhibits activity of peroxisome proliferator activated receptor α (PPAR-α)3, and lowered PPAR-α activity increased Notum expression. Genetic targeting of Notum or Wnt supplementation restored function of aged intestinal organoids. Moreover, pharmacological inhibition of Notum in mice enhanced the regenerative capacity of aged stem cells and promoted recovery from chemotherapy-induced damage. Our results reveal a role of the stem cell niche in ageing and demonstrate that targeting of Notum can promote regeneration of aged tissues.


Aging , Cellular Senescence , Esterases/metabolism , Intestinal Mucosa/pathology , Paneth Cells/metabolism , Regeneration , Aging/physiology , Animals , Cellular Senescence/physiology , Esterases/antagonists & inhibitors , Esterases/biosynthesis , Female , Humans , Intestinal Mucosa/physiology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , PPAR alpha/metabolism , Paneth Cells/pathology , Receptors, G-Protein-Coupled/metabolism , Stem Cell Niche , Stem Cells/pathology , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway
14.
Sci Rep ; 8(1): 14008, 2018 09 18.
Article En | MEDLINE | ID: mdl-30228296

Expression of the chromatin-associated protein HMGA2 correlates with progression, metastasis and therapy resistance in pancreatic ductal adenocarcinoma (PDAC). Hmga2 has also been identified as a marker of a transient subpopulation of PDAC cells that has increased metastatic ability. Here, we characterize the requirement for Hmga2 during growth, dissemination, and metastasis of PDAC in vivo using conditional inactivation of Hmga2 in well-established autochthonous mouse models of PDAC. Overall survival, primary tumour burden, presence of disseminated tumour cells in the peritoneal cavity or circulating tumour cells in the blood, and presence and number of metastases were not significantly different between mice with Hmga2-wildtype or Hmga2-deficient tumours. Treatment of mice with Hmga2-wildtype and Hmga2-deficient tumours with gemcitabine did not uncover a significant impact of Hmga2-deficiency on gemcitabine sensitivity. Hmga1 and Hmga2 overlap in their expression in both human and murine PDAC, however knockdown of Hmga1 in Hmga2-deficient cancer cells also did not decrease metastatic ability. Thus, Hmga2 remains a prognostic marker which identifies a metastatic cancer cell state in primary PDAC, however Hmga2 has limited if any direct functional impact on PDAC progression and therapy resistance.


Adenocarcinoma/secondary , Carcinoma, Pancreatic Ductal/secondary , Cell Proliferation , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , HMGA2 Protein/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Deoxycytidine/pharmacology , HMGA2 Protein/genetics , Humans , Mice , Mice, Knockout , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Prognosis , Survival Rate , Tissue Array Analysis , Tumor Cells, Cultured , Gemcitabine
15.
Nat Methods ; 14(7): 737-742, 2017 Jul.
Article En | MEDLINE | ID: mdl-28530655

Cancer growth is a multistage, stochastic evolutionary process. While cancer genome sequencing has been instrumental in identifying the genomic alterations that occur in human tumors, the consequences of these alterations on tumor growth remain largely unexplored. Conventional genetically engineered mouse models enable the study of tumor growth in vivo, but they are neither readily scalable nor sufficiently quantitative to unravel the magnitude and mode of action of many tumor-suppressor genes. Here, we present a method that integrates tumor barcoding with ultradeep barcode sequencing (Tuba-seq) to interrogate tumor-suppressor function in mouse models of human cancer. Tuba-seq uncovers genotype-dependent distributions of tumor sizes. By combining Tuba-seq with multiplexed CRISPR-Cas9-mediated genome editing, we quantified the effects of 11 tumor-suppressor pathways that are frequently altered in human lung adenocarcinoma. Tuba-seq enables the broad quantification of the function of tumor-suppressor genes with unprecedented resolution, parallelization, and precision.


Neoplasms, Experimental/metabolism , Tumor Suppressor Proteins/metabolism , Adenocarcinoma/genetics , Animals , DNA/genetics , DNA/isolation & purification , DNA/metabolism , DNA Barcoding, Taxonomic , Female , Genetic Engineering , Humans , Lentivirus/genetics , Lung/metabolism , Lung Neoplasms/genetics , Male , Mice , Models, Genetic , Plasmids , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Proteins/genetics
16.
PLoS Genet ; 11(12): e1005751, 2015 Dec.
Article En | MEDLINE | ID: mdl-26713447

Although single genes underlying several evolutionary adaptations have been identified, the genetic basis of complex, polygenic adaptations has been far more challenging to pinpoint. Here we report that the budding yeast Saccharomyces paradoxus has recently evolved resistance to citrinin, a naturally occurring mycotoxin. Applying a genome-wide test for selection on cis-regulation, we identified five genes involved in the citrinin response that are constitutively up-regulated in S. paradoxus. Four of these genes are necessary for resistance, and are also sufficient to increase the resistance of a sensitive strain when over-expressed. Moreover, cis-regulatory divergence in the promoters of these genes contributes to resistance, while exacting a cost in the absence of citrinin. Our results demonstrate how the subtle effects of individual regulatory elements can be combined, via natural selection, into a complex adaptation. Our approach can be applied to dissect the genetic basis of polygenic adaptations in a wide range of species.


Adaptation, Physiological/genetics , Genetic Fitness , Promoter Regions, Genetic , Saccharomyces/genetics , Antifungal Agents/toxicity , Citrinin/toxicity , Drug Resistance, Fungal/genetics , Genes, Fungal , Saccharomyces/drug effects , Saccharomyces/metabolism , Selection, Genetic
17.
Genes Dev ; 29(14): 1576-85, 2015 Jul 15.
Article En | MEDLINE | ID: mdl-26178787

Pancreatic ductal adenocarcinoma (PDAC) is a genomically diverse, prevalent, and almost invariably fatal malignancy. Although conventional genetically engineered mouse models of human PDAC have been instrumental in understanding pancreatic cancer development, these models are much too labor-intensive, expensive, and slow to perform the extensive molecular analyses needed to adequately understand this disease. Here we demonstrate that retrograde pancreatic ductal injection of either adenoviral-Cre or lentiviral-Cre vectors allows titratable initiation of pancreatic neoplasias that progress into invasive and metastatic PDAC. To enable in vivo CRISPR/Cas9-mediated gene inactivation in the pancreas, we generated a Cre-regulated Cas9 allele and lentiviral vectors that express Cre and a single-guide RNA. CRISPR-mediated targeting of Lkb1 in combination with oncogenic Kras expression led to selection for inactivating genomic alterations, absence of Lkb1 protein, and rapid tumor growth that phenocopied Cre-mediated genetic deletion of Lkb1. This method will transform our ability to rapidly interrogate gene function during the development of this recalcitrant cancer.


Adenocarcinoma/physiopathology , Carcinoma, Pancreatic Ductal/physiopathology , Disease Models, Animal , Adenocarcinoma/genetics , Animals , Carcinoma, Pancreatic Ductal/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Expression Regulation, Neoplastic , Genetic Vectors/genetics , Genome/genetics , Humans , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic
18.
Rev. biol. trop ; 55(1): 247-254, Mar. 2007. graf, mapas
Article Es | CUMED | ID: cum-41815

Permanently submerged mangrove roots (Rhizophora mangle) are the main habitat of the ascidian Ecteinascidia turbinata in Cuba. It was occasionally found on black coral (Antiphates caribeana) between 22 and 38 meters deep. This species exhibits a wide distribution in all the mangrove keys surrounding the Island of Cuba but does not occur in riparian or fringing mangroves. Populations of this species are abundant in Cuba: in 75 percent of the 58 localities sampled the species was present and in 57 percent more than 50 percent of the roots held at least one colony. The highest colony densities were found in the northern coast of Pinar del Rio province with values near one colony per lineal meter of mangrove root. We found the highest density (1.46 col/m) and greatest biomass at Jutías Key, with values between 25 and 660 g/m. The average of wet biomass in the studied mangroves was 73.63 g/m(AU)


Animals , Ecosystem , Plant Roots , Rhizophoraceae , Urochordata/physiology , Biomass , Cuba , Population Density , Population Dynamics , Urochordata/classification
19.
Rev. biol. trop ; 55(1): 247-254, Mar. 2007. graf, mapas
Article Es | LILACS | ID: lil-501478

Permanently submerged mangrove roots (Rhizophora mangle) are the main habitat of the ascidian Ecteinascidia turbinata in Cuba. It was occasionally found on black coral (Antiphates caribeana) between 22 and 38 meters deep. This species exhibits a wide distribution in all the mangrove keys surrounding the Island of Cuba but does not occur in riparian or fringing mangroves. Populations of this species are abundant in Cuba: in 75% of the 58 localities sampled the species was present and in 57% more than 50% of the roots held at least one colony. The highest colony densities were found in the northern coast of Pinar del Rio province with values near one colony per lineal meter of mangrove root. We found the highest density (1.46 col/m) and greatest biomass at Jutías Key, with values between 25 and 660 g/m. The average of wet biomass in the studied mangroves was 73.63 g/m.


Animals , Ecosystem , Plant Roots , Rhizophoraceae , Urochordata/physiology , Biomass , Cuba , Population Density , Population Dynamics , Urochordata/classification
20.
Rev Biol Trop ; 55(1): 247-54, 2007 Mar.
Article Es | MEDLINE | ID: mdl-18457133

Permanently submerged mangrove roots (Rhizophora mangle) are the main habitat of the ascidian Ecteinascidia turbinata in Cuba. It was occasionally found on black coral (Antiphates caribeana) between 22 and 38 meters deep. This species exhibits a wide distribution in all the mangrove keys surrounding the Island of Cuba but does not occur in riparian or fringing mangroves. Populations of this species are abundant in Cuba: in 75% of the 58 localities sampled the species was present and in 57% more than 50% of the roots held at least one colony. The highest colony densities were found in the northern coast of Pinar del Rio province with values near one colony per lineal meter of mangrove root. We found the highest density (1.46 col/m) and greatest biomass at Jutías Key, with values between 25 and 660 g/m. The average of wet biomass in the studied mangroves was 73.63 g/m.


Ecosystem , Plant Roots , Rhizophoraceae , Urochordata/physiology , Animals , Biomass , Cuba , Population Density , Population Dynamics , Urochordata/classification
...