Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Innate Immun ; 14(1): 31-41, 2022.
Article in English | MEDLINE | ID: mdl-34515137

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a very common hepatic pathology featuring steatosis and is linked to obesity and related conditions, such as the metabolic syndrome. When hepatic steatosis is accompanied by inflammation, the disorder is defined as nonalcoholic steatohepatitis (NASH), which in turn can progress toward fibrosis development that can ultimately result in cirrhosis. Cells of innate immunity, such as neutrophils or macrophages, are central regulators of NASH-related inflammation. Recent studies utilizing new experimental technologies, such as single-cell RNA sequencing, have revealed substantial heterogeneity within the macrophage populations of the liver, suggesting distinct functions of liver-resident Kupffer cells and recruited monocyte-derived macrophages with regards to regulation of liver inflammation and progression of NASH pathogenesis. Herein, we discuss recent developments concerning the function of innate immune cell subsets in NAFLD and NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Immunity, Innate , Kupffer Cells , Liver , Macrophages
2.
Hepatology ; 75(4): 881-897, 2022 04.
Article in English | MEDLINE | ID: mdl-34519101

ABSTRACT

BACKGROUND AND AIMS: NAFLD is initiated by steatosis and can progress through fibrosis and cirrhosis to HCC. The RNA binding protein human antigen R (HuR) controls RNAs at the posttranscriptional level; hepatocyte HuR has been implicated in the regulation of diet-induced hepatic steatosis. The present study aimed to understand the role of hepatocyte HuR in NAFLD development and progression to fibrosis and HCC. APPROACH AND RESULTS: Hepatocyte-specific, HuR-deficient mice and control HuR-sufficient mice were fed either a normal diet or an NAFLD-inducing diet. Hepatic lipid accumulation, inflammation, fibrosis, and HCC development were studied by histology, flow cytometry, quantitative PCR, and RNA sequencing. The liver lipidome was characterized by lipidomics analysis, and the HuR-RNA interactions in the liver were mapped by RNA immunoprecipitation sequencing. Hepatocyte-specific, HuR-deficient mice displayed spontaneous hepatic steatosis and fibrosis predisposition compared to control HuR-sufficient mice. On an NAFLD-inducing diet, hepatocyte-specific HuR deficiency resulted in exacerbated inflammation, fibrosis, and HCC-like tumor development. A multi-omic approach, including lipidomics, transcriptomics, and RNA immunoprecipitation sequencing revealed that HuR orchestrates a protective network of hepatic-metabolic and lipid homeostasis-maintaining pathways. Consistently, HuR-deficient livers accumulated, already at steady state, a triglyceride signature resembling that of NAFLD livers. Moreover, up-regulation of secreted phosphoprotein 1 expression mediated, at least partially, fibrosis development in hepatocyte-specific HuR deficiency on an NAFLD-inducing diet, as shown by experiments using antibody blockade of osteopontin. CONCLUSIONS: HuR is a gatekeeper of liver homeostasis, preventing NAFLD-related fibrosis and HCC, suggesting that the HuR-dependent network could be exploited therapeutically.


Subject(s)
Carcinoma, Hepatocellular , ELAV-Like Protein 1 , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Carcinoma, Hepatocellular/pathology , ELAV-Like Protein 1/metabolism , Homeostasis , Inflammation/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , RNA , Triglycerides/metabolism
3.
Gut ; 70(5): 940-950, 2021 05.
Article in English | MEDLINE | ID: mdl-32591434

ABSTRACT

OBJECTIVE: The rs641738C>T variant located near the membrane-bound O-acyltransferase domain containing 7 (MBOAT7) locus is associated with fibrosis in liver diseases, including non-alcoholic fatty liver disease (NAFLD), alcohol-related liver disease, hepatitis B and C. We aim to understand the mechanism by which the rs641738C>T variant contributes to pathogenesis of NAFLD. DESIGN: Mice with hepatocyte-specific deletion of MBOAT7 (Mboat7Δhep) were generated and livers were characterised by histology, flow cytometry, qPCR, RNA sequencing and lipidomics. We analysed the association of rs641738C>T genotype with liver inflammation and fibrosis in 846 NAFLD patients and obtained genotype-specific liver lipidomes from 280 human biopsies. RESULTS: Allelic imbalance analysis of heterozygous human liver samples pointed to lower expression of the MBOAT7 transcript on the rs641738C>T haplotype. Mboat7Δhep mice showed spontaneous steatosis characterised by increased hepatic cholesterol ester content after 10 weeks. After 6 weeks on a high fat, methionine-low, choline-deficient diet, mice developed increased hepatic fibrosis as measured by picrosirius staining (p<0.05), hydroxyproline content (p<0.05) and transcriptomics, while the inflammatory cell populations and inflammatory mediators were minimally affected. In a human biopsied NAFLD cohort, MBOAT7 rs641738C>T was associated with fibrosis (p=0.004) independent of the presence of histological inflammation. Liver lipidomes of Mboat7Δhep mice and human rs641738TT carriers with fibrosis showed increased total lysophosphatidylinositol levels. The altered lysophosphatidylinositol and phosphatidylinositol subspecies in MBOAT7Δhep livers and human rs641738TT carriers were similar. CONCLUSION: Mboat7 deficiency in mice and human points to an inflammation-independent pathway of liver fibrosis that may be mediated by lipid signalling and a potentially targetable treatment option in NAFLD.


Subject(s)
Acyltransferases/genetics , Liver Cirrhosis/genetics , Membrane Proteins/genetics , Non-alcoholic Fatty Liver Disease/genetics , Acyltransferases/deficiency , Adult , Aged , Animals , Biopsy , Disease Models, Animal , Disease Progression , Female , Genotype , Haplotypes , Humans , Inflammation/genetics , Male , Membrane Proteins/deficiency , Mice, Inbred C57BL , Middle Aged , Polymorphism, Single Nucleotide
4.
Cell ; 183(3): 771-785.e12, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125892

ABSTRACT

Trained innate immunity, induced via modulation of mature myeloid cells or their bone marrow progenitors, mediates sustained increased responsiveness to secondary challenges. Here, we investigated whether anti-tumor immunity can be enhanced through induction of trained immunity. Pre-treatment of mice with ß-glucan, a fungal-derived prototypical agonist of trained immunity, resulted in diminished tumor growth. The anti-tumor effect of ß-glucan-induced trained immunity was associated with transcriptomic and epigenetic rewiring of granulopoiesis and neutrophil reprogramming toward an anti-tumor phenotype; this process required type I interferon signaling irrespective of adaptive immunity in the host. Adoptive transfer of neutrophils from ß-glucan-trained mice to naive recipients suppressed tumor growth in the latter in a ROS-dependent manner. Moreover, the anti-tumor effect of ß-glucan-induced trained granulopoiesis was transmissible by bone marrow transplantation to recipient naive mice. Our findings identify a novel and therapeutically relevant anti-tumor facet of trained immunity involving appropriate rewiring of granulopoiesis.


Subject(s)
Granulocytes/immunology , Immunity, Innate , Neoplasms/immunology , Adaptive Immunity , Adoptive Transfer , Animals , Epigenesis, Genetic , Interferon Type I/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Neoplasms/pathology , Neutrophils/metabolism , Phenotype , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/metabolism , Transcription, Genetic , Transcriptome/genetics , beta-Glucans/metabolism
5.
Rev Endocr Metab Disord ; 19(4): 283-292, 2018 12.
Article in English | MEDLINE | ID: mdl-29922964

ABSTRACT

Immune cells are present in the adipose tissue (AT) and regulate its function. Under lean conditions, immune cells predominantly of type 2 immunity, including eosinophils, M2-like anti-inflammatory macrophages and innate lymphoid cells 2, contribute to the maintenance of metabolic homeostasis within the AT. In the course of obesity, pro-inflammatory immune cells, such as M1-like macrophages, prevail in the AT. Inflammation in the obese AT is associated with the development of metabolic complications such as insulin resistance, type 2 diabetes and cardiovascular disease. Thus, the immune cell-adipocyte crosstalk in the AT is an important regulator of AT function and systemic metabolism. We discuss herein this crosstalk with a special focus on the role of innate immune cells in AT inflammation and metabolic homeostasis in obesity.


Subject(s)
Adipose Tissue , Dendritic Cells , Immunity, Innate , Inflammation , Leukocytes , Macrophages , Obesity , Adipose Tissue/immunology , Adipose Tissue/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Obesity/immunology , Obesity/metabolism
6.
Nat Immunol ; 18(6): 654-664, 2017 06.
Article in English | MEDLINE | ID: mdl-28414311

ABSTRACT

In obesity, inflammation of white adipose tissue (AT) is associated with diminished generation of beige adipocytes ('beige adipogenesis'), a thermogenic and energy-dissipating function mediated by beige adipocytes that express the uncoupling protein UCP1. Here we delineated an inflammation-driven inhibitory mechanism of beige adipogenesis in obesity that required direct adhesive interactions between macrophages and adipocytes mediated by the integrin α4 and its counter-receptor VCAM-1, respectively; expression of the latter was upregulated in obesity. This adhesive interaction reciprocally and concomitantly modulated inflammatory activation of macrophages and downregulation of UCP1 expression dependent on the kinase Erk in adipocytes. Genetic or pharmacological inactivation of the integrin α4 in mice resulted in elevated expression of UCP1 and beige adipogenesis of subcutaneous AT in obesity. Our findings, established in both mouse systems and human systems, reveal a self-sustained cycle of inflammation-driven impairment of beige adipogenesis in obesity.


Subject(s)
Adipocytes, Beige , Adipogenesis/immunology , Adipose Tissue, White/immunology , Cell Differentiation/immunology , Inflammation/immunology , Macrophages/immunology , Obesity/immunology , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Adhesion/immunology , Diet, High-Fat , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback , Female , Gene Knockdown Techniques , Humans , Immunoblotting , Integrin alpha4/genetics , Macrophages/metabolism , Male , Mice , Middle Aged , Monocytes/immunology , Obesity/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Subcutaneous Fat , T-Lymphocytes/immunology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Young Adult
7.
Rev Endocr Metab Disord ; 17(1): 29-39, 2016 03.
Article in English | MEDLINE | ID: mdl-26847547

ABSTRACT

The low grade inflammatory state present in obesity promotes the progression of Non-Alcoholic Fatty Liver Disease (NAFLD). In Non-Alcoholic Steatohepatitis (NASH), augmented hepatic steatosis is accompanied by aberrant intrahepatic inflammation and exacerbated hepatocellular injury. NASH is an important disorder and can lead to fibrosis, cirrhosis and even neoplasia. The pathology of NASH involves a complex network of mechanisms, including increased infiltration of different subsets of immune cells, such as monocytes, T-lymphocytes and neutrophils, to the liver, as well as activation and in situ expansion of liver resident cells such as Kupffer cells or stellate cells. In this review, we summarize recent advances regarding understanding the role of the various cells of the innate and adaptive immunity in NASH development and progression, and discuss possible future therapeutic options and tools to interfere with disease progression.


Subject(s)
Inflammation/immunology , Metabolic Diseases/immunology , Non-alcoholic Fatty Liver Disease/immunology , Animals , Humans
8.
J Cell Physiol ; 231(7): 1534-41, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26553365

ABSTRACT

The hypoxic environment is a crucial component of the cancer stem cell niche and it is capable of eliciting stem cell features in cancer cells. We previously reported that SNAI2 up-regulates the expression of Carbonic Anhydrase iso-enzyme 9 (CA9) in hypoxic MCF7 cells. Here we show that SNAI2 down-regulates miR34a expression in hypoxic MCF7 cell-derived mammospheres. Next, we report on the capability of miR34a to decrease CA9 mRNA stability and CA9 protein expression. We also convey that the over-expression of cloned CA9-mRNA-3'UTR increases the mRNA half-life and protein levels of two miR34a targets JAGGED1 and NOTCH3. The data here reported shows that the SNAI2-dependent down-regulation of miR34a substantially contributes to the post-transcriptional up-regulation of CA9, and that CA9-mRNA-3'UTR acts as an endogenous microRNA sponge. We conclude that CA9/miR34 interplay shares in the hypoxic regulation of mammospheres and therefore, may play a relevant role in the hypoxic breast cancer stem cell niche.


Subject(s)
Antigens, Neoplasm/biosynthesis , Breast Neoplasms/genetics , Carbonic Anhydrases/biosynthesis , Cell Hypoxia/genetics , MicroRNAs/genetics , Antigens, Neoplasm/genetics , Breast Neoplasms/pathology , Calcium-Binding Proteins/biosynthesis , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/genetics , Intercellular Signaling Peptides and Proteins/biosynthesis , Jagged-1 Protein , MCF-7 Cells , Membrane Proteins/biosynthesis , MicroRNAs/biosynthesis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptor, Notch3 , Receptors, Notch/biosynthesis , Serrate-Jagged Proteins , Snail Family Transcription Factors , Transcription Factors/biosynthesis
9.
J Pharm Biomed Anal ; 112: 169-75, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-25544639

ABSTRACT

Human serum albumin (HSA) undergoes several structural alterations affecting its properties in pro-oxidant and pro-inflammatory environments, as it occurs during liver cirrhosis. These modifications include the formation of albumin dimers. Although HSA dimers were reported to be an oxidative stress biomarker, to date nothing is known about their role in liver cirrhosis and related complications. Additionally, no high sensitive analytical method was available for HSA dimers assessment in clinical settings. Thus the HSA dimeric form in human plasma was characterized by mass spectrometry using liquid chromatography tandem mass spectrometry (LC-ESI-Q-TOF) and matrix assisted laser desorption time of flight (MALDI-TOF) techniques. N-terminal and C-terminal truncated HSA, as well as the native HSA, undergo dimerization by binding another HSA molecule. This study demonstrated the presence of both homo- and hetero-dimeric forms of HSA. The dimerization site was proved to be at Cys-34, forming a disulphide bridge between two albumin molecules, as determined by LC-MS analysis after tryptic digestion. Interestingly, when plasma samples from cirrhotic subjects were analysed, the dimer/monomer ratio resulted significantly increased when compared to that of healthy subjects. These isoforms could represent promising biomarkers for liver disease. Additionally, this analytical approach leads to the relative quantification of the residual native HSA, with fully preserved structural integrity.


Subject(s)
Biomarkers/blood , Biomarkers/chemistry , Liver Cirrhosis/blood , Protein Isoforms/blood , Protein Isoforms/chemistry , Serum Albumin/chemistry , Chronic Disease , Female , Humans , Male , Middle Aged , Oxidative Stress/physiology , Protein Multimerization , Serum Albumin/biosynthesis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
10.
Am J Pathol ; 183(5): 1688-1697, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24036252

ABSTRACT

Cancer stem cell survival relies on the activation of inflammatory pathways, which is speculatively triggered by cell autonomous mechanisms or by microenvironmental stimuli. Here, we observed that hypoxic bone marrow stroma-derived transforming growth factor-ß 1 promotes the growth of human breast cancer stem cells as mammospheres. The ensuing Slug-dependent serine 139 phosphorylation of the DNA damage sensor H2AX in breast cancer stem cells induces tumor necrosis factor-α and IL-8 mRNAs, whose stability is enhanced by cytoplasmic ß-catenin. ß-Catenin also up-regulates and binds miR-221, reducing the stability of the miR-221 targets Rad51 and ERα mRNAs. Our data show that the Slug/ß-catenin-dependent activation of DNA damage signaling triggered by the hypoxic microenvironment sustains the proinflammatory phenotype of breast cancer stem cells.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Inflammation/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Transcription Factors/metabolism , beta Catenin/metabolism , Autocrine Communication/drug effects , Autocrine Communication/genetics , Breast Neoplasms/genetics , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cytokines/genetics , Cytokines/metabolism , DNA Damage/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , Humans , Inflammation/genetics , MCF-7 Cells , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Models, Biological , Neoplastic Stem Cells/drug effects , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Serine/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Snail Family Transcription Factors , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...