Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Cancer Prev Res (Phila) ; 14(12): 1089-1100, 2021 12.
Article in English | MEDLINE | ID: mdl-34389629

ABSTRACT

Mechanisms underlying aspirin chemoprevention of colorectal cancer remain unclear. Prior studies have been limited because of the inability of preclinical models to recapitulate human normal colon epithelium or cellular heterogeneity present in mucosal biopsies. To overcome some of these obstacles, we performed in vitro aspirin treatment of colon organoids derived from normal mucosal biopsies to reveal transcriptional networks relevant to aspirin chemoprevention. Colon organoids derived from 38 healthy individuals undergoing endoscopy were treated with 50 µmol/L aspirin or vehicle control for 72 hours and subjected to bulk RNA sequencing. Paired regression analysis using DESeq2 identified differentially expressed genes (DEG) associated with aspirin treatment. Cellular composition was determined using CIBERSORTx. Aspirin treatment was associated with 1,154 significant (q < 0.10) DEGs prior to deconvolution. We provide replication of these findings in an independent population-based RNA-sequencing dataset of mucosal biopsies (BarcUVa-Seq), where a significant enrichment for overlap of DEGs was observed (P < 2.2E-16). Single-cell deconvolution revealed changes in cell composition, including a decrease in transit-amplifying cells following aspirin treatment (P = 0.01). Following deconvolution, DEGs included novel putative targets for aspirin such as TRABD2A (q = 0.055), a negative regulator of Wnt signaling. Weighted gene co-expression network analysis identified 12 significant modules, including two that contained hubs for EGFR and PTGES2, the latter being previously implicated in aspirin chemoprevention. In summary, aspirin treatment of patient-derived colon organoids using physiologically relevant doses resulted in transcriptome-wide changes that reveal altered cell composition and improved understanding of transcriptional pathways, providing novel insight into its chemopreventive properties. PREVENTION RELEVANCE: Numerous studies have highlighted a role for aspirin in colorectal cancer chemoprevention, though the mechanisms driving this association remain unclear. We addressed this by showing that aspirin treatment of normal colon organoids diminished the transit-amplifying cell population, inhibited prostaglandin synthesis, and dysregulated expression of novel genes implicated in colon tumorigenesis.


Subject(s)
Organoids , Transcriptome , Aspirin/pharmacology , Colon/pathology , Humans , Sequence Analysis, RNA/methods
2.
Alcohol ; 95: 37-43, 2021 09.
Article in English | MEDLINE | ID: mdl-34118353

ABSTRACT

Identifying the minority of patients with alcohol use disorder (AUD) who develop the wide spectrum of alcohol-associated liver disease (ALD), and risk-stratifying these patients, is of critical importance. High-Mobility Group Box 1 protein (HMGB1) is an alarmin that has been implicated in the pathogenesis of multiple liver diseases. Its use as a biomarker for liver disease in those with AUD has not been studied. In this report, we investigated the association between serum HMGB1 and the presence, severity, and progression of ALD in two well-characterized cohorts of patients with AUD. In our discovery cohort of 80 patients, we found that patients with AUD and ALD exhibited higher serum HMGB1 levels compared to patients with AUD only (p = 0.0002). Additionally, serum HMGB1 levels were positively associated with liver disease severity (p < 0.0001). We found that index serum HMGB1 levels were associated with liver disease progression, defined by an increase in MELD score at 120 days (p = 0.0397). Serum HMGB1 was notable for its diagnostic and prognostic ability; it proved able to distinguish accurately between severe and non-severe forms of ALD in both our discovery cohort (AUC = 0.8199, p = 0.0003) and an independent validation cohort of 74 patients with AUD (AUC = 0.8818, p < 0.0001). Moreover, serum HMGB1 levels effectively predicted both liver-related readmission (AUC = 0.8849, p < 0.0001) and transplantation/death (AUC = 0.8614, p = 0.0002) at 90 days. The predictive potential of HMGB1 was also validated in an independent cohort of patients with AUD. Taken together, our results suggest that serum HMGB1 shows promise as a biologically relevant biomarker for ALD in patients with AUD.


Subject(s)
Alcoholism , HMGB1 Protein , Liver Diseases, Alcoholic , Alcoholism/diagnosis , Biomarkers , Humans , Liver Diseases, Alcoholic/diagnosis , Patient Readmission
4.
Methods Mol Biol ; 2265: 363-376, 2021.
Article in English | MEDLINE | ID: mdl-33704727

ABSTRACT

The lymph node microenvironment is extremely dynamic and responds to immune stimuli in the host by reprogramming immune, stromal, and endothelial cells. In normal physiological conditions, the lymph node will initiate an appropriate immune response to clear external threats that the host may experience. However, in metastatic disease, cancer cells often colonize local lymph nodes, disrupt immune function, and even leave the lymph node to create additional metastases. Understanding how cancer cells enter, colonize, survive, proliferate, and interact with other cell types in the lymph node is challenging. Here, we describe the use of photoconvertible fluorescent proteins to label and trace the fate of cancer cells once they enter the lymph node.


Subject(s)
Cell Tracking , Green Fluorescent Proteins/metabolism , Lymph Nodes , Neoplasms, Experimental , Neoplastic Cells, Circulating , Optical Imaging , Animals , Cell Line, Tumor , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Tumor Microenvironment
6.
Proc Natl Acad Sci U S A ; 117(21): 11667-11673, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32393626

ABSTRACT

Alcohol-related liver disease (ALD) accounts for the majority of cirrhosis and liver-related deaths worldwide. Activation of IFN-regulatory factor (IRF3) initiates alcohol-induced hepatocyte apoptosis, which fuels a robust secondary inflammatory response that drives ALD. The dominant molecular mechanism by which alcohol activates IRF3 and the pathways that amplify inflammatory signals in ALD remains unknown. Here we show that cytoplasmic sensor cyclic guanosine monophosphate-adenosine monophosphate (AMP) synthase (cGAS) drives IRF3 activation in both alcohol-injured hepatocytes and the neighboring parenchyma via a gap junction intercellular communication pathway. Hepatic RNA-seq analysis of patients with a wide spectrum of ALD revealed that expression of the cGAS-IRF3 pathway correlated positively with disease severity. Alcohol-fed mice demonstrated increased hepatic expression of the cGAS-IRF3 pathway. Mice genetically deficient in cGAS and IRF3 were protected against ALD. Ablation of cGAS in hepatocytes only phenocopied this hepatoprotection, highlighting the critical role of hepatocytes in fueling the cGAS-IRF3 response to alcohol. We identified connexin 32 (Cx32), the predominant hepatic gap junction, as a critical regulator of spreading cGAS-driven IRF3 activation through the liver parenchyma. Disruption of Cx32 in ALD impaired IRF3-stimulated gene expression, resulting in decreased hepatic injury despite an increase in hepatic steatosis. Taken together, these results identify cGAS and Cx32 as key factors in ALD pathogenesis and as potential therapeutic targets for hepatoprotection.


Subject(s)
Gap Junctions/metabolism , Interferon Regulatory Factor-3/metabolism , Liver Diseases, Alcoholic/metabolism , Nucleotidyltransferases/metabolism , Adult , Animals , Apoptosis , Female , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Middle Aged , Nucleotidyltransferases/genetics , Signal Transduction
7.
Nature ; 560(7717): E26, 2018 08.
Article in English | MEDLINE | ID: mdl-29849139

ABSTRACT

In Fig. 4e of this Article, the labels for 'Control' and 'HFD' were reversed ('Control' should have been labelled blue rather than purple, and 'HFD' should have been labelled purple rather than blue). Similarly, in Fig. 4f of this Article, the labels for 'V' and 'GW' were reversed ('V' should have been labelled blue rather than purple, and 'GW' should have been labelled purple instead of blue). The original figure has been corrected online.

8.
Science ; 359(6382): 1403-1407, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29567713

ABSTRACT

Lymph node metastases in cancer patients are associated with tumor aggressiveness, poorer prognoses, and the recommendation for systemic therapy. Whether cancer cells in lymph nodes can seed distant metastases has been a subject of considerable debate. We studied mice implanted with cancer cells (mammary carcinoma, squamous cell carcinoma, or melanoma) expressing the photoconvertible protein Dendra2. This technology allowed us to selectively photoconvert metastatic cells in the lymph node and trace their fate. We found that a fraction of these cells invaded lymph node blood vessels, entered the blood circulation, and colonized the lung. Thus, in mouse models, lymph node metastases can be a source of cancer cells for distant metastases. Whether this mode of dissemination occurs in cancer patients remains to be determined.


Subject(s)
Blood Vessels/pathology , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Neoplasm Seeding , Animals , Carcinoma, Squamous Cell , Cell Line, Tumor , Cell Movement , Cell Tracking/methods , Cytosol/chemistry , Female , Luminescent Proteins/analysis , Lung/pathology , Melanoma, Experimental , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplastic Cells, Circulating
10.
N Engl J Med ; 376(23): e50, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28594156
11.
Nat Biotechnol ; 35(6): 569-576, 2017 06.
Article in English | MEDLINE | ID: mdl-28459449

ABSTRACT

In vivo interrogation of the function of genes implicated in tumorigenesis is limited by the need to generate and cross germline mutant mice. Here we describe approaches to model colorectal cancer (CRC) and metastasis, which rely on in situ gene editing and orthotopic organoid transplantation in mice without cancer-predisposing mutations. Autochthonous tumor formation is induced by CRISPR-Cas9-based editing of the Apc and Trp53 tumor suppressor genes in colon epithelial cells and by orthotopic transplantation of Apc-edited colon organoids. ApcΔ/Δ;KrasG12D/+;Trp53Δ/Δ (AKP) mouse colon organoids and human CRC organoids engraft in the distal colon and metastasize to the liver. Finally, we apply the orthotopic transplantation model to characterize the clonal dynamics of Lgr5+ stem cells and demonstrate sequential activation of an oncogene in established colon adenomas. These experimental systems enable rapid in vivo characterization of cancer-associated genes and reproduce the entire spectrum of tumor progression and metastasis.


Subject(s)
Colorectal Neoplasms/genetics , Disease Models, Animal , Gene Editing/methods , Genes, Neoplasm/genetics , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Organ Transplantation/methods , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Female , Male , Mice , Mice, Transgenic , Neoplasm Metastasis
12.
Trials ; 18(1): 50, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28143522

ABSTRACT

BACKGROUND: Although aspirin is recommended for the prevention of colorectal cancer, the specific individuals for whom the benefits outweigh the risks are not clearly defined. Moreover, the precise mechanisms by which aspirin reduces the risk of cancer are unclear. We recently launched the ASPirin Intervention for the REDuction of colorectal cancer risk (ASPIRED) trial to address these uncertainties. METHODS/DESIGN: ASPIRED is a prospective, double-blind, multidose, placebo-controlled, biomarker clinical trial of aspirin use in individuals previously diagnosed with colorectal adenoma. Individuals (n = 180) will be randomized in a 1:1:1 ratio to low-dose (81 mg/day) or standard-dose (325 mg/day) aspirin or placebo. At two study visits, participants will provide lifestyle, dietary and biometric data in addition to urine, saliva and blood specimens. Stool, grossly normal colorectal mucosal biopsies and cytology brushings will be collected during a flexible sigmoidoscopy without bowel preparation. The study will examine the effect of aspirin on urinary prostaglandin metabolites (PGE-M; primary endpoint), plasma inflammatory markers (macrophage inhibitory cytokine-1 (MIC-1)), colonic expression of transcription factor binding (transcription factor 7-like 2 (TCF7L2)), colonocyte gene expression, including hydroxyprostaglandin dehydrogenase 15-(NAD) (HPGD) and those that encode Wnt signaling proteins, colonic cellular nanocytology and oral and gut microbial composition and function. DISCUSSION: Aspirin may prevent colorectal cancer through multiple, interrelated mechanisms. The ASPIRED trial will scrutinize these pathways and investigate putative mechanistically based risk-stratification biomarkers. TRIAL REGISTRATION: This protocol is registered with the U.S. National Institutes of Health trial registry, ClinicalTrials.gov, under the identifier NCT02394769 . Registered on 16 March 2015.


Subject(s)
Adenoma/drug therapy , Anticarcinogenic Agents/administration & dosage , Aspirin/administration & dosage , Carcinoma/prevention & control , Colorectal Neoplasms/drug therapy , Adenoma/metabolism , Adenoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Anticarcinogenic Agents/adverse effects , Aspirin/adverse effects , Biomarkers, Tumor/blood , Biomarkers, Tumor/urine , Boston , Carcinoma/metabolism , Carcinoma/pathology , Clinical Protocols , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytokines/blood , Disease Progression , Double-Blind Method , Female , Humans , Inflammation Mediators/blood , Male , Middle Aged , Prospective Studies , Prostaglandins/urine , Protective Factors , Research Design , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , Young Adult
13.
Clin Transl Gastroenterol ; 8(2): e73, 2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28181993

ABSTRACT

OBJECTIVES: Obesity is an important risk factor for the development of colorectal cancer (CRC). Although the impact of bariatric surgery on CRC is conflicting, its impact on precursor lesions is unknown. The aim of this study was to determine whether bariatric surgery before index screening colonoscopy is associated with decreased development of colorectal adenomas. METHODS: We performed a retrospective cohort study of bariatric surgery patients who had undergone index, screening colonoscopy at an academic center from 2001 to 2014. Patients who had bariatric surgery at least 1 year before index colonoscopy were compared with those who had surgery after colonoscopy, using multivariable logistic regression to control for presurgical body mass index, sex, gender, race, type of surgery, aspirin use, metformin use, smoking, and age at colonoscopy. RESULTS: One hundred and twenty-five obese individuals who had bariatric surgery before colonoscopy were compared with 223 individuals who had colonoscopy after surgery. Adenomatous polyps were found in 16.8% of individuals who had surgery first vs. 35.5% who had colonoscopy before bariatric surgery (unadjusted odds ratio (OR) 0.37, 95% confidence interval (CI): 0.21-0.64, P=0.0003). After multivariable adjustment, bariatric surgery before index screening colonoscopy was associated with a decreased risk of adenomas at index colonoscopy (adjusted OR 0.37, 95% CI: 0.19-0.69, P=0.002). CONCLUSIONS: Bariatric surgery is associated with a decreased risk of colorectal adenomas in obese individuals without a family history of CRC.

14.
Sci Transl Med ; 8(360): 360ra135, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27733559

ABSTRACT

The survival benefit of anti-vascular endothelial growth factor (VEGF) therapy in metastatic colorectal cancer (mCRC) patients is limited to a few months because of acquired resistance. We show that anti-VEGF therapy induced remodeling of the extracellular matrix with subsequent alteration of the physical properties of colorectal liver metastases. Preoperative treatment with bevacizumab in patients with colorectal liver metastases increased hyaluronic acid (HA) deposition within the tumors. Moreover, in two syngeneic mouse models of CRC metastasis in the liver, we show that anti-VEGF therapy markedly increased the expression of HA and sulfated glycosaminoglycans (sGAGs), without significantly changing collagen deposition. The density of these matrix components correlated with increased tumor stiffness after anti-VEGF therapy. Treatment-induced tumor hypoxia appeared to be the driving force for the remodeling of the extracellular matrix. In preclinical models, we show that enzymatic depletion of HA partially rescued the compromised perfusion in liver mCRCs after anti-VEGF therapy and prolonged survival in combination with anti-VEGF therapy and chemotherapy. These findings suggest that extracellular matrix components such as HA could be a potential therapeutic target for reducing physical barriers to systemic treatments in patients with mCRC who receive anti-VEGF therapy.


Subject(s)
Bevacizumab/therapeutic use , Colorectal Neoplasms/pathology , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Bevacizumab/adverse effects , Biomechanical Phenomena , Cell Line, Tumor , Colorectal Neoplasms/therapy , Drug Resistance, Neoplasm , Extracellular Matrix/pathology , Extracellular Matrix/physiology , Glycosaminoglycans/metabolism , Humans , Hyaluronic Acid/metabolism , Hypoxia/etiology , Hypoxia/physiopathology , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Translational Research, Biomedical
15.
Nature ; 531(7592): 53-8, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26935695

ABSTRACT

Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5(+) intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Colonic Neoplasms/pathology , Diet, High-Fat/adverse effects , Intestines/pathology , Stem Cells/drug effects , Stem Cells/pathology , Animals , Cell Count , Cell Self Renewal/drug effects , Female , Genes, APC , Humans , Male , Mice , Obesity/chemically induced , Obesity/pathology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , PPAR delta/metabolism , Signal Transduction/drug effects , Stem Cell Niche/drug effects , Stem Cells/metabolism , beta Catenin/metabolism
16.
Cancer Prev Res (Phila) ; 8(12): 1138-45, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26382604

ABSTRACT

Circulating adiponectin is inversely related to the risk of colorectal cancer. However, its influence on colorectal cancer survival is unclear. We conducted a prospective study to evaluate the association between prediagnostic plasma levels of adiponectin and mortality in patients with colorectal cancer. We identified 621 incident colorectal cancer cases who provided blood specimens prior to diagnosis within the Nurses' Health Study (NHS) and Health Professionals Follow-up Study (HPFS). Cox proportional hazards models were used to calculate HRs and 95% confidence intervals (CI). After a median follow-up of 9 years, there were 269 (43%) total deaths, of which 181 (67%) were due to colorectal cancer. Compared with participants in the lowest quartile of adiponectin, those in the highest quartile had multivariate HRs of 1.89 (95% CI, 1.21-2.97; P(trend) = 0.01) for colorectal cancer-specific mortality and 1.66 (95% CI, 1.15-2.39; P(trend) = 0.009) for overall mortality. The apparent increased risk in colorectal cancer-specific mortality was more pronounced in patients with metastatic disease (HR, 3.02: 95% CI, 1.50-6.08). Among patients with colorectal cancer, prediagnostic plasma adiponectin is associated with an increased risk of colorectal cancer-specific and overall mortality and is more apparent in patients with metastatic disease. Adiponectin may be a marker for cancers which develop through specific pathways that may be associated with worsened prognosis. Further studies are needed to validate these findings.


Subject(s)
Adiponectin/blood , Biomarkers, Tumor/blood , Colorectal Neoplasms/blood , Adult , Colorectal Neoplasms/mortality , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Survival Analysis
17.
Expert Opin Drug Discov ; 10(11): 1217-29, 2015.
Article in English | MEDLINE | ID: mdl-26295972

ABSTRACT

INTRODUCTION: Despite increased screening rates and advances in targeted therapy, colorectal cancer (CRC) remains the third leading cause of cancer-related mortality. CRC models that recapitulate key features of human disease are essential to the development of novel and effective therapeutics. Classic methods of modeling CRC such as human cell lines and xenograft mice, while useful for many applications, carry significant limitations. Recently developed in vitro and in vivo models overcome some of these deficiencies and thus can be utilized to better model CRC for mechanistic and translational research. AREAS COVERED: The authors review established models of in vitro cell culture and describe advances in organoid culture for studying normal and malignant intestine. They also discuss key features of classic xenograft models and describe other approaches for in vivo CRC research, including patient-derived xenograft, carcinogen-induced, orthotopic transplantation and transgenic mouse models. We also describe mouse models of metastatic CRC. EXPERT OPINION: No single model is optimal for drug discovery in CRC. Genetically engineered models overcome many limitations of xenograft models. Three-dimensional organoids can be efficiently derived from both normal and malignant tissue for large-scale in vitro and in vivo (transplantation) studies and are thus a significant advance in CRC drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Drug Discovery/methods , Animals , Colorectal Neoplasms/pathology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Molecular Targeted Therapy , Translational Research, Biomedical/methods , Xenograft Model Antitumor Assays
18.
20.
Mol Cancer Res ; 12(3): 464-76, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24269953

ABSTRACT

UNLABELLED: The PI3K-AKT signaling pathway regulates all phenotypes that contribute to progression of human cancers, including breast cancer. AKT mediates signal relay by phosphorylating numerous substrates, which are causally implicated in biologic responses such as cell growth, survival, metabolic reprogramming, migration, and invasion. Here a new AKT substrate is identified, the adherens junction protein Afadin, which is phosphorylated by AKT at Ser1718. Importantly, under conditions of physiologic IGF-1 signaling and oncogenic PI3K and AKT, Afadin is phosphorylated by all AKT isoforms, and this phosphorylation elicits a relocalization of Afadin from adherens junctions to the nucleus. Also, phosphorylation of Afadin increased breast cancer cell migration that was dependent on Ser1718 phosphorylation. Finally, nuclear localization of Afadin was observed in clinical breast cancer specimens, indicating that regulation of Afadin by the PI3K-AKT pathway has pathophysiologic significance. IMPLICATIONS: Phosphorylation of the adhesion protein Afadin by AKT downstream of the PI3K pathway, leads to redistribution of Afadin and controls cancer cell migration.


Subject(s)
Adherens Junctions/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/physiology , Microfilament Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adherens Junctions/genetics , Breast Neoplasms/genetics , Cell Culture Techniques , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Humans , Microfilament Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...