Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters











Publication year range
1.
STAR Protoc ; 5(3): 103303, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39264806

ABSTRACT

Applying Opal multiplex immunofluorescence (OMI) to characterize intestinal tissues of genetically engineered mouse models provides an excellent tool for studying complex processes. However, detecting appropriate signals from multiple target molecules is challenging. Here, we present a protocol to characterize mouse intestinal epithelial cell lineage using OMI. We describe steps for processing small intestine and colonic mouse tissues and designing and optimizing panels for OMI in mouse intestinal tissues. We then detail procedures for performing a quantitative evaluation of acquired images. For complete details on the use and execution of this protocol, please refer to Kinoshita et al.1.


Subject(s)
Cell Lineage , Epithelial Cells , Fluorescent Antibody Technique , Intestinal Mucosa , Animals , Mice , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fluorescent Antibody Technique/methods , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Colon/cytology , Colon/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism
2.
Dev Cell ; 59(15): 1972-1987.e8, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38815584

ABSTRACT

The early mechanisms of spontaneous tumor initiation that precede malignancy are largely unknown. We show that reduced aPKC levels correlate with stem cell loss and the induction of revival and metaplastic programs in serrated- and conventional-initiated premalignant lesions, which is perpetuated in colorectal cancers (CRCs). Acute inactivation of PKCλ/ι in vivo and in mouse organoids is sufficient to stimulate JNK in non-transformed intestinal epithelial cells (IECs), which promotes cell death and the rapid loss of the intestinal stem cells (ISCs), including those that are LGR5+. This is followed by the accumulation of revival stem cells (RSCs) at the bottom of the crypt and fetal-metaplastic cells (FMCs) at the top, creating two spatiotemporally distinct cell populations that depend on JNK-induced AP-1 and YAP. These cell lineage changes are maintained during cancer initiation and progression and determine the aggressive phenotype of human CRC, irrespective of their serrated or conventional origin.


Subject(s)
Colorectal Neoplasms , Epithelial Cells , Metaplasia , Protein Kinase C , Animals , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Protein Kinase C/metabolism , Protein Kinase C/genetics , Metaplasia/pathology , Metaplasia/metabolism , Mice , Humans , Epithelial Cells/metabolism , Epithelial Cells/pathology , Stem Cells/metabolism , Stem Cells/pathology , Transcription Factor AP-1/metabolism , Transcription Factor AP-1/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , YAP-Signaling Proteins/metabolism , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/deficiency , Organoids/metabolism , Organoids/pathology , Cell Lineage , Isoenzymes/metabolism , Isoenzymes/genetics , Isoenzymes/deficiency , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism
3.
Nat Commun ; 14(1): 8075, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38092754

ABSTRACT

The metabolic and signaling pathways regulating aggressive mesenchymal colorectal cancer (CRC) initiation and progression through the serrated route are largely unknown. Although relatively well characterized as BRAF mutant cancers, their poor response to current targeted therapy, difficult preneoplastic detection, and challenging endoscopic resection make the identification of their metabolic requirements a priority. Here, we demonstrate that the phosphorylation of SCAP by the atypical PKC (aPKC), PKCλ/ι promotes its degradation and inhibits the processing and activation of SREBP2, the master regulator of cholesterol biosynthesis. We show that the upregulation of SREBP2 and cholesterol by reduced aPKC levels is essential for controlling metaplasia and generating the most aggressive cell subpopulation in serrated tumors in mice and humans. Since these alterations are also detected prior to neoplastic transformation, together with the sensitivity of these tumors to cholesterol metabolism inhibitors, our data indicate that targeting cholesterol biosynthesis is a potential mechanism for serrated chemoprevention.


Subject(s)
Protein Kinase C , Signal Transduction , Animals , Humans , Mice , Cell Transformation, Neoplastic/genetics , Cholesterol , Epithelial Cells/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism
4.
Nat Commun ; 14(1): 5534, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37749092

ABSTRACT

Mesenchymal activation, characterized by dense stromal infiltration of immune and mesenchymal cells, fuels the aggressiveness of colorectal cancers (CRC), driving progression and metastasis. Targetable molecules in the tumor microenvironment (TME) need to be identified to improve the outcome in CRC patients with this aggressive phenotype. This study reports a positive link between high thrombospondin-1 (THBS1) expression and mesenchymal characteristics, immunosuppression, and unfavorable CRC prognosis. Bone marrow-derived monocyte-like cells recruited by CXCL12 are the primary source of THBS1, which contributes to the development of metastasis by inducing cytotoxic T-cell exhaustion and impairing vascularization. Furthermore, in orthotopically generated CRC models in male mice, THBS1 loss in the TME renders tumors partially sensitive to immune checkpoint inhibitors and anti-cancer drugs. Our study establishes THBS1 as a potential biomarker for identifying mesenchymal CRC and as a critical suppressor of antitumor immunity that contributes to the progression of this malignancy with a poor prognosis.


Subject(s)
Colorectal Neoplasms , Monocytes , Humans , Male , Animals , Mice , Immunosuppression Therapy , Aggression , Immune Checkpoint Inhibitors , Tumor Microenvironment
5.
J Gastroenterol ; 58(8): 705-717, 2023 08.
Article in English | MEDLINE | ID: mdl-37219625

ABSTRACT

The serrated neoplasia pathway constitutes an "alternative route" to colorectal cancer (CRC), and sessile serrated lesions with dysplasia (SSLDs) are an intermediate step between sessile serrated lesions (SSLs) and invasive CRC in this pathway. While SSLs show indolent growth before becoming dysplastic (> 10-15 years), SSLDs are considered to rapidly progress to either immunogenic microsatellite instable-high (MSI-H) CRC (presumably 75% of cases) or mesenchymal microsatellite stable (MSS) CRC. Their flat shapes and the relatively short window of this intermediate state make it difficult to detect and diagnose SSLDs; thus, these lesions are potent precursors of post-colonoscopy/interval cancers. Confusing terminology and the lack of longitudinal observation data of serrated polyps have hampered the accumulation of knowledge about SSLDs; however, a growing body of evidence has started to clarify their characteristics and biology. Together with recent efforts to incorporate terminology, histological studies of SSLDs have identified distinct dysplastic patterns and revealed alterations in the tumor microenvironment (TME). Molecular studies at the single-cell level have identified distinct gene alterations in both the epithelium and the TME. Mouse serrated tumor models have demonstrated the importance of TME in disease progression. Advances in colonoscopy provide clues to distinguish pre-malignant from non-malignant-SSLs. Recent progress in all aspects of the field has enhanced our understanding of the biology of SSLDs. The aim of this review article was to assess the current knowledge of SSLDs and highlight their clinical implications.


Subject(s)
Colonic Neoplasms , Colonic Polyps , Colorectal Neoplasms , Animals , Mice , Colonic Polyps/genetics , Colonic Polyps/pathology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colonoscopy , Disease Progression , Tumor Microenvironment
6.
STAR Protoc ; 4(2): 102243, 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37083323

ABSTRACT

Imaging organoid culture provides an excellent tool for studying complex diseases such as cancer. However, retaining the morphology of intact organoids for immunolabeling has been challenging. Here, we describe a protocol for immunofluorescence staining in intact colorectal cancer organoids derived from mice. We also describe additional steps for co-culture with mouse fibroblasts to enable the study of interactions with other cellular components of the tissue microenvironment. For complete details on the use and execution of this protocol, please refer to Martinez-Ordoñez et al. (2023).1.

7.
Cancer Cell ; 41(2): 252-271.e9, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36525970

ABSTRACT

Mesenchymal colorectal cancer (mCRC) is microsatellite stable (MSS), highly desmoplastic, with CD8+ T cells excluded to the stromal periphery, resistant to immunotherapy, and driven by low levels of the atypical protein kinase Cs (aPKCs) in the intestinal epithelium. We show here that a salient feature of these tumors is the accumulation of hyaluronan (HA) which, along with reduced aPKC levels, predicts poor survival. HA promotes epithelial heterogeneity and the emergence of a tumor fetal metaplastic cell (TFMC) population endowed with invasive cancer features through a network of interactions with activated fibroblasts. TFMCs are sensitive to HA deposition, and their metaplastic markers have prognostic value. We demonstrate that in vivo HA degradation with a clinical dose of hyaluronidase impairs mCRC tumorigenesis and liver metastasis and enables immune checkpoint blockade therapy by promoting the recruitment of B and CD8+ T cells, including a proportion with resident memory features, and by blocking immunosuppression.


Subject(s)
Colorectal Neoplasms , Hyaluronic Acid , Tumor Microenvironment , Humans , CD8-Positive T-Lymphocytes/pathology , Colorectal Neoplasms/pathology , Hyaluronic Acid/metabolism , Immunotherapy , Sarcoma/pathology , Tumor Microenvironment/physiology
8.
STAR Protoc ; 3(3): 101635, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36035805

ABSTRACT

Air-liquid organotypic culture models enable the study of the cellular crosstalk in the tumor microenvironment. This 3D assay recapitulates the tumor niche more faithfully than 2D culture systems and represents a versatile platform that can be easily adapted to different types of cancer cells, stromal components, or ECM composition. Here, we detail the steps to build an organotypic culture including the preparation of the organotypic structure, organotypic gels, cell seeding, gel casting, membrane processing, and image and data analysis. For complete details on the use and execution of this protocol, please refer to Linares et al. (2022).


Subject(s)
Neoplasms , Tumor Microenvironment , Cell Line, Tumor
9.
Cell Rep ; 39(6): 110792, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35545049

ABSTRACT

Reduced p62 levels are associated with the induction of the cancer-associated fibroblast (CAF) phenotype, which promotes tumorigenesis in vitro and in vivo through inflammation and metabolic reprogramming. However, how p62 is downregulated in the stroma fibroblasts by tumor cells to drive CAF activation is an unresolved central issue in the field. Here we show that tumor-secreted lactate downregulates p62 transcriptionally through a mechanism involving reduction of the NAD+/NADH ratio, which impairs poly(ADP-ribose)-polymerase 1 (PARP-1) activity. PARP-1 inhibition blocks the poly(ADP-ribosyl)ation of the AP-1 transcription factors, c-FOS and c-JUN, which is an obligate step for p62 downregulation. Importantly, restoring p62 levels in CAFs by NAD+ renders CAFs less active. PARP inhibitors, such as olaparib, mimick lactate in the reduction of stromal p62 levels, as well as the subsequent stromal activation both in vitro and in vivo, which suggests that therapies using olaparib would benefit from strategies aimed at inhibiting CAF activity.


Subject(s)
Cancer-Associated Fibroblasts , Neoplasms , Cancer-Associated Fibroblasts/metabolism , Fibroblasts/metabolism , Lactic Acid/metabolism , NAD/metabolism , Neoplasms/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism
10.
Trends Cell Biol ; 32(12): 1023-1034, 2022 12.
Article in English | MEDLINE | ID: mdl-35501226

ABSTRACT

Nononcogenic cancer drivers often impinge on complex signals that create new addictions and vulnerabilities. Protein kinase Cλ/ι (PKCλ/ι) suppresses tumorigenesis by blocking metabolic pathways that regulate fuel oxidation and create building blocks for the epigenetic control of cell differentiation. Reduced levels of PKCλ/ι unleash these pathways to promote tumorigenesis, but the simultaneous activation of the STING-driven interferon cascade prevents tumor initiation by triggering immunosurveillance mechanisms. However, depending on the context of other signaling pathways, such as WNT/ß-catenin or PKCζ, and timing, PKCλ/ι deletion can promote or inhibit tumorigenesis. In this review, we discuss in detail the molecular and cellular underpinnings of PKCλ/ι functions in cancer with the perspective of the crosstalk between metabolism and inflammation in the tumor microenvironment.


Subject(s)
Isoenzymes , Neoplasms , Humans , Isoenzymes/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Signal Transduction/physiology , Neoplasms/genetics , Cell Transformation, Neoplastic/pathology , Tumor Microenvironment
11.
J Neurosci ; 42(14): 3011-3024, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35169022

ABSTRACT

Dysregulation of autophagic pathways leads to accumulation of abnormal proteins and damaged organelles in many neurodegenerative disorders, including Parkinson's disease (PD) and Lewy body dementia (LBD). Autophagy-related dysfunction may also trigger secretion and spread of misfolded proteins, such as α-synuclein (α-syn), the major misfolded protein found in PD/LBD. However, the mechanism underlying these phenomena remains largely unknown. Here, we used cell-based models, including human induced pluripotent stem cell-derived neurons, CRISPR/Cas9 technology, and male transgenic PD/LBD mice, plus vetting in human postmortem brains (both male and female). We provide mechanistic insight into this pathologic pathway. We find that aberrant S-nitrosylation of the autophagic adaptor protein p62 causes inhibition of autophagic flux and intracellular buildup of misfolded proteins, with consequent secretion resulting in cell-to-cell spread. Thus, our data show that pathologic protein S-nitrosylation of p62 represents a critical factor not only for autophagic inhibition and demise of individual neurons, but also for α-syn release and spread of disease throughout the nervous system.SIGNIFICANCE STATEMENT In Parkinson's disease and Lewy body dementia, dysfunctional autophagy contributes to accumulation and spread of aggregated α-synuclein. Here, we provide evidence that protein S-nitrosylation of p62 inhibits autophagic flux, contributing to α-synuclein aggregation and spread.


Subject(s)
Induced Pluripotent Stem Cells , Lewy Body Disease , Parkinson Disease , RNA-Binding Proteins , alpha-Synuclein , Animals , Autophagy , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Male , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein S/metabolism , RNA-Binding Proteins/metabolism , alpha-Synuclein/metabolism
12.
Autophagy ; 18(1): 226-227, 2022 01.
Article in English | MEDLINE | ID: mdl-34895031

ABSTRACT

The mechanisms controlling immunosurveillance and immunoevasion often operate simultaneously to the triggering of the oncogenic signaling that results in tumor initiation. The resolution of the balance between anti-cancer immune responses and pro-tumorigenic pathways determines if a tumor cell survives and can remodel the microenvironment to reinforce immunosuppression or is eliminated by the immune system. Cancer cells must endure a toxic and metabolically challenging milieu. In its various forms, autophagy provides a way for transformed cells to survive by promoting catabolism and detoxification. Mounting evidence suggests that the boundaries between cancer immunity and mitogenic and metabolic programs are diffuse, with the same molecules likely serving several diverse roles in immunity and metabolism during tumor initiation and progression. Our recent data provide mechanistic detail and functional relevance of a new paradigm whereby the same signaling elements control immunity and autophagy in cancer.


Subject(s)
Interferons , Neoplasms , Autophagy/physiology , Carcinogenesis , Cell Transformation, Neoplastic , Humans , Isoenzymes/metabolism , Monitoring, Immunologic , Neoplasms/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases , Signal Transduction , Tumor Microenvironment
13.
Mol Cell ; 81(21): 4509-4526.e10, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34560002

ABSTRACT

The interferon (IFN) pathway is critical for cytotoxic T cell activation, which is central to tumor immunosurveillance and successful immunotherapy. We demonstrate here that PKCλ/ι inactivation results in the hyper-stimulation of the IFN cascade and the enhanced recruitment of CD8+ T cells that impaired the growth of intestinal tumors. PKCλ/ι directly phosphorylates and represses the activity of ULK2, promoting its degradation through an endosomal microautophagy-driven ubiquitin-dependent mechanism. Loss of PKCλ/ι results in increased levels of enzymatically active ULK2, which, by direct phosphorylation, activates TBK1 to foster the activation of the STING-mediated IFN response. PKCλ/ι inactivation also triggers autophagy, which prevents STING degradation by chaperone-mediated autophagy. Thus, PKCλ/ι is a hub regulating the IFN pathway and three autophagic mechanisms that serve to maintain its homeostatic control. Importantly, single-cell multiplex imaging and bioinformatics analysis demonstrated that low PKCλ/ι levels correlate with enhanced IFN signaling and good prognosis in colorectal cancer patients.


Subject(s)
Colorectal Neoplasms/metabolism , Interferons/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/physiology , Signal Transduction , Adult , Aged , Aged, 80 and over , Animals , Autophagy , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis , Cell Transformation, Neoplastic , Colorectal Neoplasms/mortality , Cycloheximide/chemistry , Female , HEK293 Cells , Humans , Immunophenotyping , Interferon Regulatory Factor-3/metabolism , Male , Membrane Proteins/metabolism , Mice , Middle Aged , Neoplasm Transplantation , Phosphorylation , Prognosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors , Up-Regulation
14.
Nat Commun ; 12(1): 2876, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001883

ABSTRACT

Activation of non-shivering thermogenesis is considered a promising approach to lower body weight in obesity. p62 deficiency in adipocytes reduces systemic energy expenditure but its role in sustaining mitochondrial function and thermogenesis remains unresolved. NBR1 shares a remarkable structural similarity with p62 and can interact with p62 through their respective PB1 domains. However, the physiological relevance of NBR1 in metabolism, as compared to that of p62, was not clear. Here we show that whole-body and adipocyte-specific ablation of NBR1 reverts the obesity phenotype induced by p62 deficiency by restoring global energy expenditure and thermogenesis in brown adipose tissue. Impaired adrenergic-induced browning of p62-deficient adipocytes is rescued by NBR1 inactivation, unveiling a negative role of NBR1 in thermogenesis under conditions of p62 loss. We demonstrate that upon p62 inactivation, NBR1 represses the activity of PPARγ, establishing an unexplored p62/NBR1-mediated paradigm in adipocyte thermogenesis that is critical for the control of obesity.


Subject(s)
Adipocytes/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , PPAR gamma/metabolism , Sequestosome-1 Protein/deficiency , Thermogenesis , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Animals, Newborn , Cell Nucleus/metabolism , Cells, Cultured , Energy Metabolism/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , PPAR gamma/genetics , Protein Binding , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Sequestosome-1 Protein/genetics
15.
Cancer Cell ; 39(5): 678-693.e11, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33740421

ABSTRACT

Many cancers, including pancreatic ductal adenocarcinoma (PDAC), depend on autophagy-mediated scavenging and recycling of intracellular macromolecules, suggesting that autophagy blockade should cause tumor starvation and regression. However, until now autophagy-inhibiting monotherapies have not demonstrated potent anti-cancer activity. We now show that autophagy blockade prompts established PDAC to upregulate and utilize an alternative nutrient procurement pathway: macropinocytosis (MP) that allows tumor cells to extract nutrients from extracellular sources and use them for energy generation. The autophagy to MP switch, which may be evolutionarily conserved and not cancer cell restricted, depends on activation of transcription factor NRF2 by the autophagy adaptor p62/SQSTM1. NRF2 activation by oncogenic mutations, hypoxia, and oxidative stress also results in MP upregulation. Inhibition of MP in autophagy-compromised PDAC elicits dramatic metabolic decline and regression of transplanted and autochthonous tumors, suggesting the therapeutic promise of combining autophagy and MP inhibitors in the clinic.


Subject(s)
Autophagy/physiology , Carcinoma, Pancreatic Ductal/metabolism , NF-E2-Related Factor 2/metabolism , Pancreatic Neoplasms/metabolism , Animals , Autophagy/genetics , Carcinoma, Pancreatic Ductal/immunology , Mice , NF-E2-Related Factor 2/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pancreatic Neoplasms/immunology , Pinocytosis/immunology , Pinocytosis/physiology , Sequestosome-1 Protein/metabolism , Signal Transduction/immunology , Signal Transduction/physiology , Pancreatic Neoplasms
16.
STAR Protoc ; 2(1): 100297, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33554135

ABSTRACT

In vivo interrogation of the functional role of genes implicated in colorectal cancer (CRC) is limited by the need for physiological models that mimic the disease. Here, we describe a protocol that provides the steps required for the orthotopic co-implantation of tumoral and stromal cells into the cecum and rectum to investigate the crosstalk between the tumor and its microenvironment. This protocol recapitulates metastases to the lymph nodes, liver, and lungs observed in human CRC. For complete details on the use and execution of this protocol, please refer to Kasashima et al. (2020).


Subject(s)
Cecum/metabolism , Colorectal Neoplasms/metabolism , Neoplasms, Experimental/metabolism , Rectum/metabolism , Tumor Microenvironment , Animals , Cecum/pathology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Mice , Neoplasms, Experimental/pathology , Rectum/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
17.
Dev Cell ; 56(1): 95-110.e10, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33207226

ABSTRACT

Cancer-associated fibroblasts (CAFs) promote tumor malignancy, but the precise transcriptional mechanisms regulating the acquisition of the CAF phenotype are not well understood. We show that the upregulation of SOX2 is central to this process, which is repressed by protein kinase Cζ (PKCζ). PKCζ deficiency activates the reprogramming of colonic fibroblasts to generate a predominant SOX2-dependent CAF population expressing the WNT regulator Sfrp2 as its top biomarker. SOX2 directly binds the Sfrp1/2 promoters, and the inactivation of Sox2 or Sfrp1/2 in CAFs impaired the induction of migration and invasion of colon cancer cells, as well as their tumorigenicity in vivo. Importantly, recurrence-free and overall survival of colorectal cancer (CRC) patients negatively correlates with stromal PKCζ levels. Also, SOX2 expression in the stroma is associated with CRC T invasion and worse prognosis of recurrence-free survival. Therefore, the PKCζ-SOX2 axis emerges as a critical step in the control of CAF pro-tumorigenic potential.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinogenesis/metabolism , Colorectal Neoplasms/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Protein Kinase C/deficiency , SOXB1 Transcription Factors/metabolism , Animals , Cancer-Associated Fibroblasts/pathology , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Invasiveness/genetics , Organoids/metabolism , Organoids/pathology , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA-Seq , Recurrence , SOXB1 Transcription Factors/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Single-Cell Analysis , Up-Regulation , beta Catenin/genetics , beta Catenin/metabolism
18.
STAR Protoc ; 1(3): 100185, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377079

ABSTRACT

This protocol provides the steps required for a mouse liver orthotopic implantation model. The reliable pre-clinical animal models that have similar characteristics to hepatocellular carcinoma (HCC) are a powerful tool to unveil the mechanisms controlling tumor initiation and progression. Here, we describe a syngeneic orthotopic HCC model that recapitulates the role of a host pro-tumorigenic microenvironment by pre-conditioning mouse livers with a high-fat diet (HFD). For complete details on the use and execution of this protocol, please refer to Kudo et al. (2020).


Subject(s)
Carcinoma, Hepatocellular/pathology , Fatty Liver/complications , Liver Neoplasms/pathology , Animals , Disease Models, Animal , Liver/pathology , Mice, Inbred C57BL , Sutures
20.
Nat Metab ; 2(10): 1034-1045, 2020 10.
Article in English | MEDLINE | ID: mdl-32839596

ABSTRACT

Benign hepatosteatosis, affected by lipid uptake, de novo lipogenesis and fatty acid (FA) oxidation, progresses to non-alcoholic steatohepatitis (NASH) on stress and inflammation. A key macronutrient proposed to increase hepatosteatosis and NASH risk is fructose. Excessive intake of fructose causes intestinal-barrier deterioration and endotoxaemia. However, how fructose triggers these alterations and their roles in hepatosteatosis and NASH pathogenesis remain unknown. Here we show, using mice, that microbiota-derived Toll-like receptor (TLR) agonists promote hepatosteatosis without affecting fructose-1-phosphate (F1P) and cytosolic acetyl-CoA. Activation of mucosal-regenerative gp130 signalling, administration of the YAP-induced matricellular protein CCN1 or expression of the antimicrobial peptide Reg3b (beta) peptide counteract fructose-induced barrier deterioration, which depends on endoplasmic-reticulum stress and subsequent endotoxaemia. Endotoxin engages TLR4 to trigger TNF production by liver macrophages, thereby inducing lipogenic enzymes that convert F1P and acetyl-CoA to FA in both mouse and human hepatocytes.


Subject(s)
Fructose/pharmacology , Inflammation/metabolism , Lipogenesis/drug effects , Acetyl Coenzyme A/pharmacology , Animals , Endotoxemia/blood , Female , Fructosephosphates/pharmacology , Gastrointestinal Microbiome , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Intestines/drug effects , Lipidomics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Regeneration/drug effects , Toll-Like Receptors/agonists
SELECTION OF CITATIONS
SEARCH DETAIL