Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 167(4): 1052-1066.e18, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27814504

ABSTRACT

It is widely believed that inflammation associated with obesity has an important role in the development of type 2 diabetes. IκB kinase beta (IKKß) is a crucial kinase that responds to inflammatory stimuli such as tumor necrosis factor α (TNF-α) by initiating a variety of intracellular signaling cascades and is considered to be a key element in the inflammation-mediated development of insulin resistance. We show here, contrary to expectation, that IKKß-mediated inflammation is a positive regulator of hepatic glucose homeostasis. IKKß phosphorylates the spliced form of X-Box Binding Protein 1 (XBP1s) and increases the activity of XBP1s. We have used three experimental approaches to enhance the IKKß activity in the liver of obese mice and observed increased XBP1s activity, reduced ER stress, and a significant improvement in insulin sensitivity and consequently in glucose homeostasis. Our results reveal a beneficial role of IKKß-mediated hepatic inflammation in glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress , Glucose/metabolism , I-kappa B Kinase/metabolism , X-Box Binding Protein 1/metabolism , Animals , Cell Line, Tumor , Homeostasis , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Phosphorylation , Protein Stability
2.
Cell ; 162(4): 766-79, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26276631

ABSTRACT

Compensatory proliferation triggered by hepatocyte loss is required for liver regeneration and maintenance but also promotes development of hepatocellular carcinoma (HCC). Despite extensive investigation, the cells responsible for hepatocyte restoration or HCC development remain poorly characterized. We used genetic lineage tracing to identify cells responsible for hepatocyte replenishment following chronic liver injury and queried their roles in three distinct HCC models. We found that a pre-existing population of periportal hepatocytes, located in the portal triads of healthy livers and expressing low amounts of Sox9 and other bile-duct-enriched genes, undergo extensive proliferation and replenish liver mass after chronic hepatocyte-depleting injuries. Despite their high regenerative potential, these so-called hybrid hepatocytes do not give rise to HCC in chronically injured livers and thus represent a unique way to restore tissue function and avoid tumorigenesis. This specialized set of pre-existing differentiated cells may be highly suitable for cell-based therapy of chronic hepatocyte-depleting disorders.


Subject(s)
Hepatocytes/transplantation , Liver/cytology , Liver/physiology , Animals , Bile Ducts/cytology , Cell Proliferation , Cell Transplantation/methods , Hepatocytes/classification , Hepatocytes/cytology , Liver/injuries , Liver Neoplasms , Mice , Regeneration , SOX9 Transcription Factor/genetics , Transcriptome
3.
Nat Immunol ; 16(12): 1235-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26502405

ABSTRACT

Ectopic lymphoid-like structures (ELSs) are often observed in cancer, yet their function is obscure. Although ELSs signify good prognosis in certain malignancies, we found that hepatic ELSs indicated poor prognosis for hepatocellular carcinoma (HCC). We studied an HCC mouse model that displayed abundant ELSs and found that they constituted immunopathological microniches wherein malignant hepatocyte progenitor cells appeared and thrived in a complex cellular and cytokine milieu until gaining self-sufficiency. The egress of progenitor cells and tumor formation were associated with the autocrine production of cytokines previously provided by the niche. ELSs developed via cooperation between the innate immune system and adaptive immune system, an event facilitated by activation of the transcription factor NF-κB and abolished by depletion of T cells. Such aberrant immunological foci might represent new targets for cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , Lymphoid Tissue/immunology , Neoplastic Stem Cells/immunology , Stem Cell Niche/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Comparative Genomic Hybridization , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Immunity, Innate/genetics , Immunity, Innate/immunology , Immunoblotting , In Situ Hybridization , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Niche/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcriptome/genetics , Transcriptome/immunology
4.
Am J Pathol ; 194(6): 1033-1046, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38423355

ABSTRACT

Low-grade chronic inflammation contributes to both aging and the pathogenesis of age-related diseases. White adipose tissue (WAT) in obese individuals exhibits chronic inflammation, which is associated with obesity-related disorders. Aging exacerbates obesity-related inflammation in WAT; however, the molecular mechanisms underlying chronic inflammation and its exacerbation by aging remain unclear. Age-related decline in activity of the proteasome, a multisubunit proteolytic complex, has been implicated in age-related diseases. This study employed a mouse model with decreased proteasomal function that exhibits age-related phenotypes to investigate the impact of adipocyte senescence on WAT inflammation. Transgenic mice expressing proteasomal subunit ß5t with weak chymotrypsin-like activity experience reduced lifespan and develop age-related phenotypes. Mice fed with a high-fat diet and experiencing proteasomal dysfunction exhibited increased WAT inflammation, increased infiltration of proinflammatory M1-like macrophages, and increased proinflammatory adipocytokine-like monocyte chemoattractant protein-1, plasminogen activator inhibitor-1, and tumor necrosis factor-α, which are all associated with activation of endoplasmic reticulum (ER) stress-related pathways. Impaired proteasomal activity also activated ER stress-related molecules and induced expression of proinflammatory adipocytokines in adipocyte-like cells differentiated from 3T3-L1 cells. Collectively, the results suggesed that impaired proteasomal activity increases ER stress and that subsequent inflammatory pathways play pivotal roles in WAT inflammation. Because proteasomal function declines with age, age-related proteasome impairment may be involved in obesity-related inflammation among elderly individuals.


Subject(s)
Endoplasmic Reticulum Stress , Inflammation , Mice, Transgenic , Obesity , Proteasome Endopeptidase Complex , Animals , Proteasome Endopeptidase Complex/metabolism , Endoplasmic Reticulum Stress/physiology , Inflammation/pathology , Inflammation/metabolism , Obesity/metabolism , Obesity/pathology , Mice , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Mice, Inbred C57BL , Diet, High-Fat/adverse effects , Adipocytes/metabolism , Adipocytes/pathology , Male , Macrophages/metabolism , Macrophages/pathology , Aging/pathology , Aging/metabolism , Adipose Tissue/pathology , Adipose Tissue/metabolism , 3T3-L1 Cells , Chronic Disease
5.
Liver Int ; 44(4): 1011-1023, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38293713

ABSTRACT

BACKGROUND & AIMS: Recently, the association between hepatocellular carcinoma (HCC) and ferroptosis has been the focus of much attention. The expression of long chain fatty acyl-CoA ligase 4 (ACSL4), a marker of ferroptosis, in tumour tissue is related to better prognosis in various cancers. In HCC, ACSL4 expression indicates poor prognosis and is related to high malignancy. However, the mechanism remains to be fully understood. METHODS: We retrospectively enrolled 358 patients with HCC who had undergone hepatic resection. Immunohistochemistry (IHC) for ACSL4 was performed. Factors associated with ASCL4 expression were investigated by spatial transcriptome analysis, and the relationships were investigated by IHC. The association between ACSL4 and the tumour immune microenvironment was examined in a public dataset and investigated by IHC. RESULTS: Patients were divided into ACSL4-positive (n = 72, 20.1%) and ACSL4-negative (n = 286, 79.9%) groups. ACSL4 positivity was significantly correlated with higher α-fetoprotein (p = .0180) and more histological liver fibrosis (p = .0014). In multivariate analysis, ACSL4 positivity was an independent prognostic factor (p < .0001). Spatial transcriptome analysis showed a positive correlation between ACSL4 and cancer-associated fibroblasts; this relationship was confirmed by IHC. Evaluation of a public dataset showed the correlation between ACSL4 and exhausted tumour immune microenvironment; this relationship was also confirmed by IHC. CONCLUSION: ACSL4 is a prognostic factor in HCC patients and its expression was associated with cancer-associated fibroblasts and anti-tumour immunity.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/surgery , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Retrospective Studies , Prognosis , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Tumor Microenvironment
6.
Gastroenterology ; 163(5): 1391-1406.e24, 2022 11.
Article in English | MEDLINE | ID: mdl-35963362

ABSTRACT

BACKGROUND & AIMS: In the mouse intestinal epithelium, Lgr5+ stem cells are vulnerable to injury, owing to their predominantly cycling nature, and their progenies de-differentiate to replenish the stem cell pool. However, how human colonic stem cells behave in homeostasis and during regeneration remains unknown. METHODS: Transcriptional heterogeneity among colonic epithelial cells was analyzed by means of single-cell RNA sequencing analysis of human and mouse colonic epithelial cells. To trace the fate of human colonic stem or differentiated cells, we generated LGR5-tdTomato, LGR5-iCasase9-tdTomato, LGR5-split-Cre, and KRT20-ERCreER knock-in human colon organoids via genome engineering. p27+ dormant cells were further visualized with the p27-mVenus reporter. To analyze the dynamics of human colonic stem cells in vivo, we orthotopically xenotransplanted fluorescence-labeled human colon organoids into immune-deficient mice. The cell cycle dynamics in xenograft cells were evaluated using 5-ethynyl-2'-deoxyuridine pulse-chase analysis. The clonogenic capacity of slow-cycling human stem cells or differentiated cells was analyzed in the context of homeostasis, LGR5 ablation, and 5-fluorouracil-induced mucosal injury. RESULTS: Single-cell RNA sequencing analysis illuminated the presence of nondividing LGR5+ stem cells in the human colon. Visualization and lineage tracing of slow-cycling LGR5+p27+ cells and orthotopic xenotransplantation validated their homeostatic lineage-forming capability in vivo, which was augmented by 5-FU-induced mucosal damage. Transforming growth factor-ß signaling regulated the quiescent state of LGR5+ cells. Despite the plasticity of differentiated KRT20+ cells, they did not display clonal growth after 5-FU-induced injury, suggesting that occupation of the niche environment by LGR5+p27+ cells prevented neighboring differentiated cells from de-differentiating. CONCLUSIONS: Our results highlight the quiescent nature of human LGR5+ colonic stem cells and their contribution to post-injury regeneration.


Subject(s)
Receptors, G-Protein-Coupled , Stem Cells , Humans , Mice , Animals , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Colon/metabolism , Intestinal Mucosa/metabolism , Fluorouracil , Transforming Growth Factors/metabolism
7.
Immunity ; 41(6): 1052-63, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25526314

ABSTRACT

Interleukin-17A (IL-17A) is a pro-inflammatory cytokine linked to rapid malignant progression of colorectal cancer (CRC) and therapy resistance. IL-17A exerts its pro-tumorigenic activity through its type A receptor (IL-17RA). However, IL-17RA is expressed in many cell types, including hematopoietic, fibroblastoid, and epithelial cells, in the tumor microenvironment, and how IL-17RA engagement promotes colonic tumorigenesis is unknown. Here we show that IL-17RA signals directly within transformed colonic epithelial cells (enterocytes) to promote early tumor development. IL-17RA engagement activates ERK, p38 MAPK, and NF-κB signaling and promotes the proliferation of tumorigenic enterocytes that just lost expression of the APC tumor suppressor. Although IL-17RA signaling also controls the production of IL-6, this mechanism makes only a partial contribution to colonic tumorigenesis. Combined treatment with chemotherapy, which induces IL-17A expression, and an IL-17A neutralizing antibody enhanced the therapeutic responsiveness of established colon tumors. These findings establish IL-17A and IL-17RA as therapeutic targets in colorectal cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/immunology , Colorectal Neoplasms/immunology , Enterocytes/physiology , Receptors, Interleukin-17/metabolism , Aberrant Crypt Foci/genetics , Animals , Antibodies, Blocking/administration & dosage , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line, Transformed , Colonic Neoplasms/chemically induced , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/drug therapy , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Enterocytes/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorouracil/administration & dosage , Humans , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Tamoxifen/administration & dosage , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Genes Dev ; 29(12): 1271-84, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26109050

ABSTRACT

YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif) are major downstream effectors of the Hippo pathway that influences tissue homeostasis, organ size, and cancer development. Aberrant hyperactivation of YAP/TAZ causes tissue overgrowth and tumorigenesis, whereas their inactivation impairs tissue development and regeneration. Dynamic and precise control of YAP/TAZ activity is thus important to ensure proper physiological regulation and homeostasis of the cells. Here, we show that YAP/TAZ activation results in activation of their negative regulators, LATS1/2 (large tumor suppressor 1/2) kinases, to constitute a negative feedback loop of the Hippo pathway in both cultured cells and mouse tissues. YAP/TAZ in complex with the transcription factor TEAD (TEA domain family member) directly induce LATS2 expression. Furthermore, YAP/TAZ also stimulate the kinase activity of LATS1/2 through inducing NF2 (neurofibromin 2). This feedback regulation is responsible for the transient activation of YAP upon lysophosphatidic acid (LPA) stimulation and the inhibition of YAP-induced cell migration. Thus, this LATS-mediated feedback loop provides an efficient mechanism to establish the robustness and homeostasis of YAP/TAZ regulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Feedback, Physiological/physiology , Homeostasis/physiology , Neurofibromin 2/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Movement , Cells, Cultured , DNA-Binding Proteins/metabolism , Enzyme Activation/physiology , Female , Gene Expression Regulation , HEK293 Cells , Hippo Signaling Pathway , Homeostasis/genetics , Humans , Liver/metabolism , Male , Mice , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
9.
Nature ; 519(7541): 57-62, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25731159

ABSTRACT

Inflammation promotes regeneration of injured tissues through poorly understood mechanisms, some of which involve interleukin (IL)-6 family members, the expression of which is elevated in many diseases including inflammatory bowel diseases and colorectal cancer. Here we show in mice and human cells that gp130, a co-receptor for IL-6 cytokines, triggers activation of YAP and Notch, transcriptional regulators that control tissue growth and regeneration, independently of the gp130 effector STAT3. Through YAP and Notch, intestinal gp130 signalling stimulates epithelial cell proliferation, causes aberrant differentiation and confers resistance to mucosal erosion. gp130 associates with the related tyrosine kinases Src and Yes, which are activated on receptor engagement to phosphorylate YAP and induce its stabilization and nuclear translocation. This signalling module is strongly activated upon mucosal injury to promote healing and maintain barrier function.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytokine Receptor gp130/metabolism , Epithelial Cells/cytology , Inflammation/metabolism , Intestinal Mucosa/cytology , Phosphoproteins/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Regeneration , Animals , Body Weight , Cell Cycle Proteins , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Enzyme Activation , Epithelial Cells/metabolism , Epithelial Cells/pathology , HEK293 Cells , Homeostasis , Humans , Inflammation/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Proto-Oncogene Proteins c-yes/metabolism , Receptors, Notch/metabolism , Signal Transduction , Up-Regulation , YAP-Signaling Proteins
10.
Proc Natl Acad Sci U S A ; 115(42): E9879-E9888, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30287485

ABSTRACT

Cancer genomics has enabled the exhaustive molecular characterization of tumors and exposed hepatocellular carcinoma (HCC) as among the most complex cancers. This complexity is paralleled by dozens of mouse models that generate histologically similar tumors but have not been systematically validated at the molecular level. Accurate models of the molecular pathogenesis of HCC are essential for biomedical progress; therefore we compared genomic and transcriptomic profiles of four separate mouse models [MUP transgenic, TAK1-knockout, carcinogen-driven diethylnitrosamine (DEN), and Stelic Animal Model (STAM)] with those of 987 HCC patients with distinct etiologies. These four models differed substantially in their mutational load, mutational signatures, affected genes and pathways, and transcriptomes. STAM tumors were most molecularly similar to human HCC, with frequent mutations in Ctnnb1, similar pathway alterations, and high transcriptomic similarity to high-grade, proliferative human tumors with poor prognosis. In contrast, TAK1 tumors better reflected the mutational signature of human HCC and were transcriptionally similar to low-grade human tumors. DEN tumors were least similar to human disease and almost universally carried the Braf V637E mutation, which is rarely found in human HCC. Immune analysis revealed that strain-specific MHC-I genotype can influence the molecular makeup of murine tumors. Thus, different mouse models of HCC recapitulate distinct aspects of HCC biology, and their use should be adapted to specific questions based on the molecular features provided here.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Gene Expression Profiling , Genomics/methods , Liver Neoplasms, Experimental/genetics , Liver Neoplasms/genetics , Animals , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Humans , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Transcriptome
11.
Int J Mol Sci ; 22(5)2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33668122

ABSTRACT

Chronic inflammation is thought to promote tumorigenesis and metastasis by several mechanisms, such as affecting tumor cells directly, establishing a tumor-supporting microenvironment, enhancing tumor angiogenesis, and suppressing antitumor immunity. In this review, we discuss the recent advances in our understanding of how inflammation induces the immunosuppressive tumor microenvironment, such as increasing the level of pro-inflammatory cytokines, chemokines, and immunosuppressive molecules, inducing immune checkpoint molecules and cytotoxic T-cell exhaustion, and accumulating regulatory T (Treg) cells and myeloid-derived suppressor cells (MDSCs). The suppression of antitumor immunity by inflammation is especially examined in the liver and colorectal cancer. In addition, chronic inflammation is induced during aging and causes age-related diseases, including cancer, by affecting immunity. Therefore, we also discuss the age-related diseases regulated by inflammation, especially in the liver and colon.


Subject(s)
Aging/immunology , Cell Transformation, Neoplastic/immunology , Gastrointestinal Neoplasms/immunology , Immunomodulation , Inflammation/immunology , Liver Neoplasms/immunology , Aging/pathology , Animals , Cell Transformation, Neoplastic/pathology , Gastrointestinal Neoplasms/pathology , Humans , Liver Neoplasms/pathology
12.
Int J Mol Sci ; 22(11)2021 May 21.
Article in English | MEDLINE | ID: mdl-34063828

ABSTRACT

Inflammation, especially chronic inflammation, plays a pivotal role in tumorigenesis and metastasis through various mechanisms and is now recognized as a hallmark of cancer and an attractive therapeutic target in cancer. In this review, we discuss recent advances in molecular mechanisms of how inflammation promotes tumorigenesis and metastasis and suppresses anti-tumor immunity in various types of solid tumors, including esophageal, gastric, colorectal, liver, and pancreatic cancer as well as hematopoietic malignancies.


Subject(s)
Carcinogenesis/pathology , Inflammation/pathology , Neoplasm Metastasis/pathology , Animals , Hematologic Neoplasms/pathology , Humans
13.
J Hepatol ; 72(6): 1182-1195, 2020 06.
Article in English | MEDLINE | ID: mdl-32105670

ABSTRACT

BACKGROUND & AIMS: Hepatomegaly can be triggered by insulin and insulin-unrelated etiologies. Insulin acts via AKT, but how other challenges cause hepatomegaly is unknown. METHODS: Since many hepatomegaly-inducing toxicants and stressors activate NRF2, we examined the effect of NRF2 activation on liver size and metabolism using a conditional allele encoding a constitutively active NRF2 variant to generate Nrf2Act-hep mice in which NRF2 is selectively activated in hepatocytes. We also used adenoviruses encoding variants of the autophagy adaptor p62/SQSTM1, which activates liver NRF2, as well as liver-specific ATG7-deficient mice (Atg7Δhep) and liver specimens from patients with hepatic sinusoidal obstruction syndrome (HSOS) and autoimmune hepatitis (AIH). RNA sequencing and cell signaling analyses were used to determine cellular consequences of NRF2 activation and diverse histological analyses were used to study effects of the different manipulations on liver and systemic pathophysiology. RESULTS: Hepatocyte-specific NRF2 activation, due to p62 accumulation or inhibition of KEAP1 binding, led to hepatomegaly associated with enhanced glycogenosis, steatosis and G2/M cell cycle arrest, fostering hyperplasia without cell division. Surprisingly, all manipulations that led to NRF2 activation also activated AKT, whose inhibition blocked NRF2-induced hepatomegaly and glycogenosis, but not NRF2-dependent antioxidant gene induction. AKT activation was linked to NRF2-mediated transcriptional induction of PDGF and EGF receptor ligands that signaled through their cognate receptors in an autocrine manner. Insulin and insulin-like growth factors were not involved. The NRF2-AKT signaling axis was also activated in human HSOS- and AIH-related hepatomegaly. CONCLUSIONS: NRF2, a transcription factor readily activated by xenobiotics, oxidative stress and autophagy disruptors, may be a common mediator of hepatomegaly; its effects on hepatic metabolism can be reversed by AKT/tyrosine kinase inhibitors. LAY SUMMARY: Hepatomegaly can be triggered by numerous etiological factors, including infections, liver cancer, metabolic disturbances, toxicant exposure, as well as alcohol abuse or drug-induced hepatitis. This study identified the oxidative stress response transcription factor NRF2 as a common mediator of hepatomegaly. NRF2 activation results in elevated expression of several growth factors. These growth factors are made by hepatocytes and activate their receptors in an autocrine fashion to stimulate the accumulation of glycogen and lipids that lead to hepatocyte and liver enlargement. The protein kinase AKT plays a key role in this process and its inhibition leads to reversal of hepatomegaly.


Subject(s)
ErbB Receptors/metabolism , Genes, erbB-1 , Hepatic Veno-Occlusive Disease/complications , Hepatic Veno-Occlusive Disease/metabolism , Hepatitis, Autoimmune/complications , Hepatitis, Autoimmune/metabolism , Hepatomegaly/complications , Hepatomegaly/metabolism , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adult , Animals , Autophagy/genetics , Disease Models, Animal , ErbB Receptors/genetics , Female , Hemangioma/metabolism , Hemangioma/pathology , Hepatic Veno-Occlusive Disease/pathology , Hepatitis, Autoimmune/pathology , Hepatomegaly/genetics , Hepatomegaly/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/genetics
14.
Proc Natl Acad Sci U S A ; 114(7): 1643-1648, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28130546

ABSTRACT

Loss of tumor suppressor adenomatous polyposis coli (APC) activates ß-catenin to initiate colorectal tumorigenesis. However, ß-catenin (CTNNB1) activating mutations rarely occur in human colorectal cancer (CRC). We found that APC loss also results in up-regulation of IL-6 signal transducer (IL-6ST/gp130), thereby activating Src family kinases (SFKs), YAP, and STAT3, which are simultaneously up-regulated in the majority of human CRC. Although, initial YAP activation, which stimulates IL6ST gene transcription, may be caused by reduced serine phosphorylation, sustained YAP activation depends on tyrosine phosphorylation by SFKs, whose inhibition, along with STAT3-activating JAK kinases, causes regression of established colorectal tumors. These results explain why APC loss is a more potent initiating event than the mere activation of CTNNB1.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Colorectal Neoplasms/metabolism , Cytokine Receptor gp130/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adult , Aged , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cytokine Receptor gp130/genetics , Female , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Mutation , beta Catenin/genetics , beta Catenin/metabolism
15.
Cancer Sci ; 110(5): 1525-1535, 2019 May.
Article in English | MEDLINE | ID: mdl-30874331

ABSTRACT

The Ras/Raf/ERK pathway is one of the most frequently dysregulated signaling pathways in various cancers. In some such cancers, Ras and Raf are hotspots for mutations, which cause continuous activation of this pathway. However, in some other cancers, it is known that negative regulators of the Ras/Raf/ERK pathway are responsible for uncontrolled activation. The Sprouty/Spred family is broadly recognized as important negative regulators of the Ras/Raf/ERK pathway, and its expression is downregulated in many malignancies, leading to hyperactivation of the Ras/Raf/ERK pathway. After the discovery of this family, intensive research investigated the mechanism by which it suppresses the Ras/Raf/ERK pathway and its roles in developmental and pathophysiological processes. In this review, we discuss the complicated roles of the Sprouty/Spred family in tumor initiation, promotion, and progression and its future therapeutic potential.


Subject(s)
Membrane Proteins/metabolism , Neoplasms/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Multigene Family , ras Proteins/metabolism
16.
Nature ; 491(7423): 254-8, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23034650

ABSTRACT

Approximately 2% of colorectal cancer is linked to pre-existing inflammation known as colitis-associated cancer, but most develops in patients without underlying inflammatory bowel disease. Colorectal cancer often follows a genetic pathway whereby loss of the adenomatous polyposis coli (APC) tumour suppressor and activation of ß-catenin are followed by mutations in K-Ras, PIK3CA and TP53, as the tumour emerges and progresses. Curiously, however, 'inflammatory signature' genes characteristic of colitis-associated cancer are also upregulated in colorectal cancer. Further, like most solid tumours, colorectal cancer exhibits immune/inflammatory infiltrates, referred to as 'tumour-elicited inflammation'. Although infiltrating CD4(+) T(H)1 cells and CD8(+) cytotoxic T cells constitute a positive prognostic sign in colorectal cancer, myeloid cells and T-helper interleukin (IL)-17-producing (T(H)17) cells promote tumorigenesis, and a 'T(H)17 expression signature' in stage I/II colorectal cancer is associated with a drastic decrease in disease-free survival. Despite its pathogenic importance, the mechanisms responsible for the appearance of tumour-elicited inflammation are poorly understood. Many epithelial cancers develop proximally to microbial communities, which are physically separated from immune cells by an epithelial barrier. We investigated mechanisms responsible for tumour-elicited inflammation in a mouse model of colorectal tumorigenesis, which, like human colorectal cancer, exhibits upregulation of IL-23 and IL-17. Here we show that IL-23 signalling promotes tumour growth and progression, and development of a tumoural IL-17 response. IL-23 is mainly produced by tumour-associated myeloid cells that are likely to be activated by microbial products, which penetrate the tumours but not adjacent tissue. Both early and late colorectal neoplasms exhibit defective expression of several barrier proteins. We propose that barrier deterioration induced by colorectal-cancer-initiating genetic lesions results in adenoma invasion by microbial products that trigger tumour-elicited inflammation, which in turn drives tumour growth.


Subject(s)
Adenoma/microbiology , Adenoma/pathology , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Interleukin-17/immunology , Interleukin-23/immunology , Adenoma/genetics , Adenoma/immunology , Animals , Bacteria/metabolism , Bacteria/pathogenicity , Cell Division , Colitis/complications , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Disease Models, Animal , Disease-Free Survival , Genes, APC , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Interleukin-17/genetics , Interleukin-23/deficiency , Interleukin-23/genetics , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Tumor Microenvironment , beta Catenin/metabolism
17.
Semin Immunol ; 26(1): 54-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24552665

ABSTRACT

Inflammatory responses play pivotal roles in cancer development, including tumor initiation, promotion, progression, and metastasis. Cytokines are now recognized as important mediators linking inflammation and cancer, and are therefore potential therapeutic and preventive targets as well as prognostic factors. The interleukin (IL)-6 family of cytokines, especially IL-6 and IL-11, is highly up-regulated in many cancers and considered as one of the most important cytokine families during tumorigenesis and metastasis. This review discusses molecular mechanisms linking the IL-6 cytokine family to solid malignancies and their treatment.


Subject(s)
Cytokines/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Neoplasms/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cytokines/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Interleukin-6/genetics , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neovascularization, Pathologic , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/metabolism , Signal Transduction/drug effects
18.
Proc Natl Acad Sci U S A ; 111(48): 17200-5, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25404284

ABSTRACT

Triclosan [5-chloro-2-(2,4-dichlorophenoxy)phenol; TCS] is a synthetic, broad-spectrum antibacterial chemical used in a wide range of consumer products including soaps, cosmetics, therapeutics, and plastics. The general population is exposed to TCS because of its prevalence in a variety of daily care products as well as through waterborne contamination. TCS is linked to a multitude of health and environmental effects, ranging from endocrine disruption and impaired muscle contraction to effects on aquatic ecosystems. We discovered that TCS was capable of stimulating liver cell proliferation and fibrotic responses, accompanied by signs of oxidative stress. Through a reporter screening assay with an array of nuclear xenobiotic receptors (XenoRs), we found that TCS activates the nuclear receptor constitutive androstane receptor (CAR) and, contrary to previous reports, has no significant effect on mouse peroxisome proliferation activating receptor α (PPARα). Using the procarcinogen diethylnitrosamine (DEN) to initiate tumorigenesis in mice, we discovered that TCS substantially accelerates hepatocellular carcinoma (HCC) development, acting as a liver tumor promoter. TCS-treated mice exhibited a large increase in tumor multiplicity, size, and incidence compared with control mice. TCS-mediated liver regeneration and fibrosis preceded HCC development and may constitute the primary tumor-promoting mechanism through which TCS acts. These findings strongly suggest there are adverse health effects in mice with long-term TCS exposure, especially on enhancing liver fibrogenesis and tumorigenesis, and the relevance of TCS liver toxicity to humans should be evaluated.


Subject(s)
Carcinogenesis/drug effects , Carcinoma, Hepatocellular/chemically induced , Liver Neoplasms/chemically induced , Liver/drug effects , Triclosan/toxicity , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Animals , Anti-Infective Agents, Local/toxicity , Cell Line , Cell Proliferation/drug effects , Cytokines/genetics , Cytokines/metabolism , Fibrosis/chemically induced , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Immunohistochemistry , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
Proc Natl Acad Sci U S A ; 111(3): 1090-5, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395807

ABSTRACT

E-cadherin is an important adhesion molecule whose loss is associated with progression and poor prognosis of liver cancer. However, it is unclear whether the loss of E-cadherin is a real culprit or a bystander in liver cancer progression. In addition, the precise role of E-cadherin in maintaining liver homeostasis is also still unknown, especially in vivo. Here we demonstrate that liver-specific E-cadherin knockout mice develop spontaneous periportal inflammation via an impaired intrahepatic biliary network, as well as periductal fibrosis, which resembles primary sclerosing cholangitis. Inducible gene knockout studies identified E-cadherin loss in biliary epithelial cells as a causal factor of cholangitis induction. Furthermore, a few of the E-cadherin knockout mice developed spontaneous liver cancer. When knockout of E-cadherin is combined with Ras activation or chemical carcinogen administration, E-cadherin knockout mice display markedly accelerated carcinogenesis and an invasive phenotype associated with epithelial-mesenchymal transition, up-regulation of stem cell markers, and elevated ERK activation. Also in human hepatocellular carcinoma, E-cadherin loss correlates with increased expression of mesenchymal and stem cell markers, and silencing of E-cadherin in hepatocellular carcinoma cell lines causes epithelial-mesenchymal transition and increased invasiveness, suggesting that E-cadherin loss can be a causal factor of these phenotypes. Thus, E-cadherin plays critical roles in maintaining homeostasis and suppressing carcinogenesis in the liver.


Subject(s)
Cadherins/metabolism , Carcinogenesis , Cholangitis, Sclerosing/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Animals , Bacterial Proteins/metabolism , Cholangitis/metabolism , Epithelial-Mesenchymal Transition , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatocytes/cytology , Inflammation , Liver/pathology , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neoplasm Metastasis , Phenotype , Prognosis , Stem Cells/cytology
20.
Nat Genet ; 39(9): 1120-6, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17704776

ABSTRACT

We report germline loss-of-function mutations in SPRED1 in a newly identified autosomal dominant human disorder. SPRED1 is a member of the SPROUTY/SPRED family of proteins that act as negative regulators of RAS->RAF interaction and mitogen-activated protein kinase (MAPK) signaling. The clinical features of the reported disorder resemble those of neurofibromatosis type 1 and consist of multiple café-au-lait spots, axillary freckling and macrocephaly. Melanocytes from a café-au-lait spot showed, in addition to the germline SPRED1 mutation, an acquired somatic mutation in the wild-type SPRED1 allele, indicating that complete SPRED1 inactivation is needed to generate a café-au-lait spot in this syndrome. This disorder is yet another member of the recently characterized group of phenotypically overlapping syndromes caused by mutations in the genes encoding key components of the RAS-MAPK pathway. To our knowledge, this is the first report of mutations in the SPRY (SPROUTY)/SPRED family of genes in human disease.


Subject(s)
Germ-Line Mutation , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Aged , Analysis of Variance , Cell Line , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Immunoblotting , Infant , Intracellular Signaling Peptides and Proteins/physiology , Male , Membrane Proteins/physiology , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Neurofibromatosis 1/metabolism , Pedigree , Phenotype , Signal Transduction/genetics , Signal Transduction/physiology , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL