Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94.125
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 375-399, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38360545

ABSTRACT

The liver's unique characteristics have a profound impact on the priming and maintenance of adaptive immunity. This review delves into the cellular circuits that regulate adaptive immune responses in the liver, with a specific focus on hepatitis B virus infection as an illustrative example. A key aspect highlighted is the liver's specialized role in priming CD8+ T cells, leading to a distinct state of immune hyporesponsiveness. Additionally, the influence of the liver's hemodynamics and anatomical features, particularly during liver fibrosis and cirrhosis, on the differentiation and function of adaptive immune cells is discussed. While the primary emphasis is on CD8+ T cells, recent findings regarding the involvement of B cells and CD4+ T cells in hepatic immunity are also reviewed. Furthermore, we address the challenges ahead and propose integrating cutting-edge techniques, such as spatial biology, and combining mouse models with human sample analyses to gain comprehensive insights into the liver's adaptive immunity. This understanding could pave the way for novel therapeutic strategies targeting infectious diseases, malignancies, and inflammatory liver conditions like metabolic dysfunction-associated steatohepatitis and autoimmune hepatitis.


Subject(s)
Adaptive Immunity , Liver , Humans , Animals , Liver/immunology , Liver/metabolism , Liver/pathology , CD8-Positive T-Lymphocytes/immunology , Hepatitis B virus/immunology , Hepatitis B virus/physiology , Hepatitis B/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology
2.
Annu Rev Immunol ; 38: 649-671, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32040356

ABSTRACT

A plethora of experimental and epidemiological evidence supports a critical role for inflammation and adaptive immunity in the onset of cancer and in shaping its response to therapy. These data are particularly robust for gastrointestinal (GI) cancers, such as those affecting the GI tract, liver, and pancreas, on which this review is focused. We propose a unifying hypothesis according to which intestinal barrier disruption is the origin of tumor-promoting inflammation that acts in conjunction with tissue-specific cancer-initiating mutations. The gut microbiota and its products impact tissue-resident and recruited myeloid cells that promote tumorigenesis through secretion of growth- and survival-promoting cytokines that act on epithelial cells, as well as fibrogenic and immunosuppressive cytokines that interfere with the proper function of adaptive antitumor immunity. Understanding these relationships should improve our ability to prevent cancer development and stimulate the immune system to eliminate existing malignancies.


Subject(s)
Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastrointestinal Microbiome , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/metabolism , Host-Pathogen Interactions/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Adaptive Immunity , Animals , Gastric Mucosa/pathology , Gastrointestinal Microbiome/immunology , Gastrointestinal Neoplasms/pathology , Humans , Immunity, Innate , Intestinal Mucosa/pathology , Liver/immunology , Liver/metabolism , Liver/pathology
3.
Cell ; 185(2): 250-265.e16, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35021064

ABSTRACT

Methods to deliver gene editing agents in vivo as ribonucleoproteins could offer safety advantages over nucleic acid delivery approaches. We report the development and application of engineered DNA-free virus-like particles (eVLPs) that efficiently package and deliver base editor or Cas9 ribonucleoproteins. By engineering VLPs to overcome cargo packaging, release, and localization bottlenecks, we developed fourth-generation eVLPs that mediate efficient base editing in several primary mouse and human cell types. Using different glycoproteins in eVLPs alters their cellular tropism. Single injections of eVLPs into mice support therapeutic levels of base editing in multiple tissues, reducing serum Pcsk9 levels 78% following 63% liver editing, and partially restoring visual function in a mouse model of genetic blindness. In vitro and in vivo off-target editing from eVLPs was virtually undetected, an improvement over AAV or plasmid delivery. These results establish eVLPs as promising vehicles for therapeutic macromolecule delivery that combine key advantages of both viral and nonviral delivery.


Subject(s)
Drug Delivery Systems , Genetic Engineering , Proteins/therapeutic use , Virion/genetics , Animals , Base Sequence , Blindness/genetics , Blindness/therapy , Brain/metabolism , DNA/metabolism , Disease Models, Animal , Fibroblasts/metabolism , Gene Editing , HEK293 Cells , Humans , Liver/pathology , Mice , Mice, Inbred C57BL , Proprotein Convertase 9/metabolism , Retinal Pigment Epithelium/pathology , Retroviridae , Virion/ultrastructure , Vision, Ocular
4.
Cell ; 184(10): 2537-2564, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33989548

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the leading chronic liver disease worldwide. Its more advanced subtype, nonalcoholic steatohepatitis (NASH), connotes progressive liver injury that can lead to cirrhosis and hepatocellular carcinoma. Here we provide an in-depth discussion of the underlying pathogenetic mechanisms that lead to progressive liver injury, including the metabolic origins of NAFLD, the effect of NAFLD on hepatic glucose and lipid metabolism, bile acid toxicity, macrophage dysfunction, and hepatic stellate cell activation, and consider the role of genetic, epigenetic, and environmental factors that promote fibrosis progression and risk of hepatocellular carcinoma in NASH.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Carcinoma, Hepatocellular/pathology , Humans , Liver/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/pathology
5.
Cell ; 184(22): 5559-5576.e19, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34678143

ABSTRACT

Glucose consumption is generally increased in tumor cells to support tumor growth. Interestingly, we report that glycogen accumulation is a key initiating oncogenic event during liver malignant transformation. We found that glucose-6-phosphatase (G6PC) catalyzing the last step of glycogenolysis is frequently downregulated to augment glucose storage in pre-malignant cells. Accumulated glycogen undergoes liquid-liquid phase separation, which results in the assembly of the Laforin-Mst1/2 complex and consequently sequesters Hippo kinases Mst1/2 in glycogen liquid droplets to relieve their inhibition on Yap. Moreover, G6PC or another glycogenolysis enzyme-liver glycogen phosphorylase (PYGL) deficiency in both human and mice results in glycogen storage disease along with liver enlargement and tumorigenesis in a Yap-dependent manner. Consistently, elimination of glycogen accumulation abrogates liver growth and cancer incidence, whereas increasing glycogen storage accelerates tumorigenesis. Thus, we concluded that cancer-initiating cells adapt a glycogen storing mode, which blocks Hippo signaling through glycogen phase separation to augment tumor incidence.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Glycogen/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Cell Line , Disease Models, Animal , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Glucose-6-Phosphatase/metabolism , Glycogen Phosphorylase/metabolism , Hepatocyte Growth Factor/metabolism , Hippo Signaling Pathway , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasm Staging , Phase Transition , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proto-Oncogene Proteins/metabolism , Serine-Threonine Kinase 3/metabolism , YAP-Signaling Proteins/metabolism
6.
Cell ; 183(6): 1536-1550.e17, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33306954

ABSTRACT

Hendra (HeV) and Nipah (NiV) viruses are emerging zoonotic pathogens in the Henipavirus genus causing outbreaks of disease with very high case fatality rates. Here, we report the first naturally occurring human monoclonal antibodies (mAbs) against HeV receptor binding protein (RBP). All isolated mAbs neutralized HeV, and some also neutralized NiV. Epitope binning experiments identified five major antigenic sites on HeV-RBP. Animal studies demonstrated that the most potent cross-reactive neutralizing mAbs, HENV-26 and HENV-32, protected ferrets in lethal models of infection with NiV Bangladesh 3 days after exposure. We solved the crystal structures of mAb HENV-26 in complex with both HeV-RBP and NiV-RBP and of mAb HENV-32 in complex with HeV-RBP. The studies reveal diverse sites of vulnerability on RBP recognized by potent human mAbs that inhibit virus by multiple mechanisms. These studies identify promising prophylactic antibodies and define protective epitopes that can be used in rational vaccine design.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hendra Virus/immunology , Henipavirus/immunology , Neutralization Tests , Nipah Virus/immunology , Receptors, Virus/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Antigens, Viral/immunology , Binding Sites , Binding, Competitive , Brain/pathology , Chiroptera/virology , Cross Reactions/immunology , Crystallography, X-Ray , Ephrin-B2/metabolism , Female , Ferrets/virology , Humans , Interferometry , Liver/pathology , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Receptors, Virus/chemistry , Receptors, Virus/metabolism
7.
Cell ; 183(1): 76-93.e22, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32931733

ABSTRACT

Mitochondria, which play central roles in immunometabolic diseases, have their own genome. However, the functions of mitochondria-located noncoding RNAs are largely unknown due to the absence of a specific delivery system. By circular RNA (circRNA) expression profile analysis of liver fibroblasts from patients with nonalcoholic steatohepatitis (NASH), we observe that mitochondrial circRNAs account for a considerable fraction of downregulated circRNAs in NASH fibroblasts. By constructing mitochondria-targeting nanoparticles, we observe that Steatohepatitis-associated circRNA ATP5B Regulator (SCAR), which is located in mitochondria, inhibits mitochondrial ROS (mROS) output and fibroblast activation. circRNA SCAR, mediated by PGC-1α, binds to ATP5B and shuts down mPTP by blocking CypD-mPTP interaction. Lipid overload inhibits PGC-1α by endoplasmic reticulum (ER) stress-induced CHOP. In vivo, targeting circRNA SCAR alleviates high fat diet-induced cirrhosis and insulin resistance. Clinically, circRNA SCAR is associated with steatosis-to-NASH progression. Collectively, we identify a mitochondrial circRNA that drives metaflammation and serves as a therapeutic target for NASH.


Subject(s)
Mitochondria/genetics , Mitochondrial Proton-Translocating ATPases/genetics , RNA, Circular/genetics , Animals , Cell Line , Diet, High-Fat , Endoplasmic Reticulum Stress/physiology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression/genetics , Humans , Insulin Resistance , Liver/pathology , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RNA, Circular/metabolism , Reactive Oxygen Species , Transcriptome/genetics
8.
Cell ; 183(2): 377-394.e21, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32976798

ABSTRACT

We employed scRNA sequencing to extensively characterize the cellular landscape of human liver from development to disease. Analysis of ∼212,000 cells representing human fetal, hepatocellular carcinoma (HCC), and mouse liver revealed remarkable fetal-like reprogramming of the tumor microenvironment. Specifically, the HCC ecosystem displayed features reminiscent of fetal development, including re-emergence of fetal-associated endothelial cells (PLVAP/VEGFR2) and fetal-like (FOLR2) tumor-associated macrophages. In a cross-species comparative analysis, we discovered remarkable similarity between mouse embryonic, fetal-liver, and tumor macrophages. Spatial transcriptomics further revealed a shared onco-fetal ecosystem between fetal liver and HCC. Furthermore, gene regulatory analysis, spatial transcriptomics, and in vitro functional assays implicated VEGF and NOTCH signaling in maintaining onco-fetal ecosystem. Taken together, we report a shared immunosuppressive onco-fetal ecosystem in fetal liver and HCC. Our results unravel a previously unexplored onco-fetal reprogramming of the tumor ecosystem, provide novel targets for therapeutic interventions in HCC, and open avenues for identifying similar paradigms in other cancers and disease.


Subject(s)
Carcinoma, Hepatocellular/pathology , Endothelial Cells/metabolism , Tumor Microenvironment/genetics , Adult , Animals , Carcinoma, Hepatocellular/genetics , Cell Line , Disease Models, Animal , Endothelial Cells/pathology , Female , Folate Receptor 2/metabolism , Gene Expression Profiling/methods , Humans , Liver/pathology , Liver Neoplasms/genetics , Macrophages/metabolism , Male , Membrane Proteins/metabolism , Mice , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics , Transcriptome/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Cell ; 183(7): 1867-1883.e26, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33248023

ABSTRACT

Biliary atresia (BA) is a severe cholangiopathy that leads to liver failure in infants, but its pathogenesis remains to be fully characterized. By single-cell RNA profiling, we observed macrophage hypo-inflammation, Kupffer cell scavenger function defects, cytotoxic T cell expansion, and deficiency of CX3CR1+effector T and natural killer (NK) cells in infants with BA. More importantly, we discovered that hepatic B cell lymphopoiesis did not cease after birth and that tolerance defects contributed to immunoglobulin G (IgG)-autoantibody accumulation in BA. In a rhesus-rotavirus induced BA model, depleting B cells or blocking antigen presentation ameliorated liver damage. In a pilot clinical study, we demonstrated that rituximab was effective in depleting hepatic B cells and restoring the functions of macrophages, Kupffer cells, and T cells to levels comparable to those of control subjects. In summary, our comprehensive immune profiling in infants with BA had educed that B-cell-modifying therapies may alleviate liver pathology.


Subject(s)
Biliary Atresia/immunology , Biliary Atresia/therapy , Liver/immunology , Animals , Antigens, CD20/metabolism , B-Lymphocytes/immunology , Biliary Atresia/blood , Biliary Atresia/drug therapy , Biopsy , CX3C Chemokine Receptor 1/metabolism , Cell Death , Cell Line , Cell Proliferation , Cell Transdifferentiation , Child , Child, Preschool , Cohort Studies , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Humans , Immunoglobulin G/metabolism , Infant , Inflammation/pathology , Killer Cells, Natural/immunology , Kupffer Cells/pathology , Liver/pathology , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Lymphocyte Depletion , Lymphopoiesis , Male , Mice, Inbred BALB C , Phagocytosis , RNA/metabolism , Rituximab/administration & dosage , Rituximab/pharmacology , Rituximab/therapeutic use , Rotavirus/physiology , Single-Cell Analysis , Th1 Cells/immunology , Th17 Cells/immunology
10.
Cell ; 177(4): 881-895.e17, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31051106

ABSTRACT

Non-alcoholic fatty liver is the most common liver disease worldwide. Here, we show that the mitochondrial protein mitofusin 2 (Mfn2) protects against liver disease. Reduced Mfn2 expression was detected in liver biopsies from patients with non-alcoholic steatohepatitis (NASH). Moreover, reduced Mfn2 levels were detected in mouse models of steatosis or NASH, and its re-expression in a NASH mouse model ameliorated the disease. Liver-specific ablation of Mfn2 in mice provoked inflammation, triglyceride accumulation, fibrosis, and liver cancer. We demonstrate that Mfn2 binds phosphatidylserine (PS) and can specifically extract PS into membrane domains, favoring PS transfer to mitochondria and mitochondrial phosphatidylethanolamine (PE) synthesis. Consequently, hepatic Mfn2 deficiency reduces PS transfer and phospholipid synthesis, leading to endoplasmic reticulum (ER) stress and the development of a NASH-like phenotype and liver cancer. Ablation of Mfn2 in liver reveals that disruption of ER-mitochondrial PS transfer is a new mechanism involved in the development of liver disease.


Subject(s)
GTP Phosphohydrolases/metabolism , Mitochondrial Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Phosphatidylserines/metabolism , Animals , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation/metabolism , Liver/pathology , Liver Diseases/etiology , Liver Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Primary Cell Culture , Protein Transport/physiology , Signal Transduction , Triglycerides/metabolism
11.
Cell ; 178(6): 1478-1492.e20, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31474362

ABSTRACT

Liver fibrosis is a very common condition seen in millions of patients with various liver diseases, and yet no effective treatments are available owing to poorly characterized molecular pathogenesis. Here, we show that leukocyte cell-derived chemotaxin 2 (LECT2) is a functional ligand of Tie1, a poorly characterized endothelial cell (EC)-specific orphan receptor. Upon binding to Tie1, LECT2 interrupts Tie1/Tie2 heterodimerization, facilitates Tie2/Tie2 homodimerization, activates PPAR signaling, and inhibits the migration and tube formations of EC. In vivo studies showed that LECT2 overexpression inhibits portal angiogenesis, promotes sinusoid capillarization, and worsens fibrosis, whereas these changes were reversed in Lect2-KO mice. Adeno-associated viral vector serotype 9 (AAV9)-LECT2 small hairpin RNA (shRNA) treatment significantly attenuates fibrosis. Upregulation of LECT2 is associated with advanced human liver fibrosis staging. We concluded that targeting LECT2/Tie1 signaling may represent a potential therapeutic target for liver fibrosis, and serum LECT2 level may be a potential biomarker for the screening and diagnosis of liver fibrosis.


Subject(s)
Endothelial Cells/metabolism , Hepatocytes/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Liver Cirrhosis/metabolism , Liver/metabolism , Receptors, TIE/metabolism , Animals , Biomarkers/metabolism , Capillaries/metabolism , Endothelial Cells/cytology , Endothelial Cells/pathology , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/pathology , Humans , Intercellular Signaling Peptides and Proteins/blood , Liver/blood supply , Liver/pathology , Liver Cirrhosis/diagnosis , Mice, Inbred C57BL
12.
Cell ; 177(3): 608-621.e12, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30955891

ABSTRACT

Normal tissues accumulate genetic changes with age, but it is unknown if somatic mutations promote clonal expansion of non-malignant cells in the setting of chronic degenerative diseases. Exome sequencing of diseased liver samples from 82 patients revealed a complex mutational landscape in cirrhosis. Additional ultra-deep sequencing identified recurrent mutations in PKD1, PPARGC1B, KMT2D, and ARID1A. The number and size of mutant clones increased as a function of fibrosis stage and tissue damage. To interrogate the functional impact of mutated genes, a pooled in vivo CRISPR screening approach was established. In agreement with sequencing results, examination of 147 genes again revealed that loss of Pkd1, Kmt2d, and Arid1a promoted clonal expansion. Conditional heterozygous deletion of these genes in mice was also hepatoprotective in injury assays. Pre-malignant somatic alterations are often viewed through the lens of cancer, but we show that mutations can promote regeneration, likely independent of carcinogenesis.


Subject(s)
Liver Diseases/pathology , Liver/metabolism , Regeneration , Animals , Chronic Disease , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Hydrolases/deficiency , Hydrolases/genetics , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Diseases/genetics , Male , Mice , Mice, Knockout , Middle Aged , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Regeneration/physiology , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Exome Sequencing
13.
Cell ; 179(4): 829-845.e20, 2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31675496

ABSTRACT

The immune microenvironment of hepatocellular carcinoma (HCC) is poorly characterized. Combining two single-cell RNA sequencing technologies, we produced transcriptomes of CD45+ immune cells for HCC patients from five immune-relevant sites: tumor, adjacent liver, hepatic lymph node (LN), blood, and ascites. A cluster of LAMP3+ dendritic cells (DCs) appeared to be the mature form of conventional DCs and possessed the potential to migrate from tumors to LNs. LAMP3+ DCs also expressed diverse immune-relevant ligands and exhibited potential to regulate multiple subtypes of lymphocytes. Of the macrophages in tumors that exhibited distinct transcriptional states, tumor-associated macrophages (TAMs) were associated with poor prognosis, and we established the inflammatory role of SLC40A1 and GPNMB in these cells. Further, myeloid and lymphoid cells in ascites were predominantly linked to tumor and blood origins, respectively. The dynamic properties of diverse CD45+ cell types revealed by this study add new dimensions to the immune landscape of HCC.


Subject(s)
Carcinoma, Hepatocellular/immunology , Cation Transport Proteins/genetics , Inflammation/immunology , Liver Neoplasms/immunology , Membrane Glycoproteins/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Communication/genetics , Cell Communication/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Inflammation/genetics , Inflammation/pathology , Leukocyte Common Antigens/immunology , Liver/immunology , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocytes/immunology , Lymphocytes/pathology , Lysosomal Membrane Proteins/genetics , Macrophages/immunology , Macrophages/pathology , Myeloid Cells/immunology , Myeloid Cells/pathology , Neoplasm Proteins/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome/genetics , Transcriptome/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
14.
Immunity ; 57(10): 2310-2327.e6, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39317200

ABSTRACT

The liver macrophage population comprises resident Kupffer cells (KCs) and monocyte-derived macrophages with distinct pro- or anti-inflammatory properties that affect the severity and course of liver diseases. The mechanisms underlying macrophage differentiation and functions in metabolic dysfunction-associated steatotic liver disease and/or steatohepatitis (MASLD/MASH) remain mostly unknown. Using single-cell RNA sequencing (scRNA-seq) and fate mapping of hepatic macrophage subpopulations, we unraveled the temporal and spatial dynamics of distinct monocyte and monocyte-derived macrophage subsets in MASH. We revealed a crucial role for the Notch-Recombination signal binding protein for immunoglobulin kappa J region (RBPJ) signaling pathway in controlling the monocyte-to-macrophage transition, with Rbpj deficiency blunting inflammatory macrophages and monocyte-derived KC differentiation and conversely promoting the emergence of protective Ly6Clo monocytes. Mechanistically, Rbpj deficiency promoted lipid uptake driven by elevated CD36 expression in Ly6Clo monocytes, enhancing their protective interactions with endothelial cells. Our findings uncover the crucial role of Notch-RBPJ signaling in monocyte-to-macrophage transition and will aid in the design of therapeutic strategies for MASH treatment.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein , Inflammation , Macrophages , Receptors, Notch , Signal Transduction , Animals , Receptors, Notch/metabolism , Mice , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Macrophages/immunology , Macrophages/metabolism , Inflammation/immunology , Inflammation/metabolism , Fatty Liver/metabolism , Fatty Liver/immunology , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Cell Differentiation , Kupffer Cells/metabolism , Kupffer Cells/immunology , Mice, Knockout , Humans , Liver/metabolism , Liver/pathology
15.
Nat Rev Mol Cell Biol ; 22(11): 751-771, 2021 11.
Article in English | MEDLINE | ID: mdl-34285405

ABSTRACT

Insulin resistance, defined as a defect in insulin-mediated control of glucose metabolism in tissues - prominently in muscle, fat and liver - is one of the earliest manifestations of a constellation of human diseases that includes type 2 diabetes and cardiovascular disease. These diseases are typically associated with intertwined metabolic abnormalities, including obesity, hyperinsulinaemia, hyperglycaemia and hyperlipidaemia. Insulin resistance is caused by a combination of genetic and environmental factors. Recent genetic and biochemical studies suggest a key role for adipose tissue in the development of insulin resistance, potentially by releasing lipids and other circulating factors that promote insulin resistance in other organs. These extracellular factors perturb the intracellular concentration of a range of intermediates, including ceramide and other lipids, leading to defects in responsiveness of cells to insulin. Such intermediates may cause insulin resistance by inhibiting one or more of the proximal components in the signalling cascade downstream of insulin (insulin receptor, insulin receptor substrate (IRS) proteins or AKT). However, there is now evidence to support the view that insulin resistance is a heterogeneous disorder that may variably arise in a range of metabolic tissues and that the mechanism for this effect likely involves a unified insulin resistance pathway that affects a distal step in the insulin action pathway that is more closely linked to the terminal biological response. Identifying these targets is of major importance, as it will reveal potential new targets for treatments of diseases associated with insulin resistance.


Subject(s)
Antigens, CD/genetics , Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Insulin/genetics , Receptor, Insulin/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Glucose/genetics , Glucose/metabolism , Humans , Insulin/metabolism , Liver/metabolism , Liver/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics
16.
Cell ; 175(5): 1289-1306.e20, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30454647

ABSTRACT

Obesity is a major driver of cancer, especially hepatocellular carcinoma (HCC). The prevailing view is that non-alcoholic steatohepatitis (NASH) and fibrosis or cirrhosis are required for HCC in obesity. Here, we report that NASH and fibrosis and HCC in obesity can be dissociated. We show that the oxidative hepatic environment in obesity inactivates the STAT-1 and STAT-3 phosphatase T cell protein tyrosine phosphatase (TCPTP) and increases STAT-1 and STAT-3 signaling. TCPTP deletion in hepatocytes promoted T cell recruitment and ensuing NASH and fibrosis as well as HCC in obese C57BL/6 mice that normally do not develop NASH and fibrosis or HCC. Attenuating the enhanced STAT-1 signaling prevented T cell recruitment and NASH and fibrosis but did not prevent HCC. By contrast, correcting STAT-3 signaling prevented HCC without affecting NASH and fibrosis. TCPTP-deletion in hepatocytes also markedly accelerated HCC in mice treated with a chemical carcinogen that promotes HCC without NASH and fibrosis. Our studies reveal how obesity-associated hepatic oxidative stress can independently contribute to the pathogenesis of NASH, fibrosis, and HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/pathology , Obesity/pathology , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Hepatocellular/metabolism , Diet, High-Fat , Disease Models, Animal , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Oxidative Stress , Protein Tyrosine Phosphatase, Non-Receptor Type 2/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Signal Transduction
17.
Immunity ; 56(1): 58-77.e11, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36521495

ABSTRACT

Obesity-induced chronic liver inflammation is a hallmark of nonalcoholic steatohepatitis (NASH)-an aggressive form of nonalcoholic fatty liver disease. However, it remains unclear how such a low-grade, yet persistent, inflammation is sustained in the liver. Here, we show that the macrophage phagocytic receptor TREM2, induced by hepatocyte-derived sphingosine-1-phosphate, was required for efferocytosis of lipid-laden apoptotic hepatocytes and thereby maintained liver immune homeostasis. However, prolonged hypernutrition led to the production of proinflammatory cytokines TNF and IL-1ß in the liver to induce TREM2 shedding through ADAM17-dependent proteolytic cleavage. Loss of TREM2 resulted in aberrant accumulation of dying hepatocytes, thereby further augmenting proinflammatory cytokine production. This ultimately precipitated a vicious cycle that licensed chronic inflammation to drive simple steatosis transition to NASH. Therefore, impaired macrophage efferocytosis is a previously unrecognized key pathogenic event that enables chronic liver inflammation in obesity. Blocking TREM2 cleavage to restore efferocytosis may represent an effective strategy to treat NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Overnutrition , Humans , Non-alcoholic Fatty Liver Disease/pathology , Overnutrition/pathology , Liver/pathology , Inflammation/pathology , Obesity/pathology , Membrane Glycoproteins , Receptors, Immunologic
18.
Cell ; 170(4): 664-677.e11, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28802039

ABSTRACT

The process of aging and circadian rhythms are intimately intertwined, but how peripheral clocks involved in metabolic homeostasis contribute to aging remains unknown. Importantly, caloric restriction (CR) extends lifespan in several organisms and rewires circadian metabolism. Using young versus old mice, fed ad libitum or under CR, we reveal reprogramming of the circadian transcriptome in the liver. These age-dependent changes occur in a highly tissue-specific manner, as demonstrated by comparing circadian gene expression in the liver versus epidermal and skeletal muscle stem cells. Moreover, de novo oscillating genes under CR show an enrichment in SIRT1 targets in the liver. This is accompanied by distinct circadian hepatic signatures in NAD+-related metabolites and cyclic global protein acetylation. Strikingly, this oscillation in acetylation is absent in old mice while CR robustly rescues global protein acetylation. Our findings indicate that the clock operates at the crossroad between protein acetylation, liver metabolism, and aging.


Subject(s)
Aging/metabolism , Circadian Rhythm , Liver/metabolism , Metabolic Networks and Pathways , Acetyl Coenzyme A/metabolism , Acetylation , Aging/pathology , Animals , Caloric Restriction , Histones/metabolism , Liver/pathology , Mice , NAD/metabolism , Proteins/metabolism , Sirtuin 1/metabolism , Stem Cells/metabolism , Transcriptome
19.
Annu Rev Cell Dev Biol ; 34: 333-355, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30028641

ABSTRACT

Stellate cells are resident lipid-storing cells of the pancreas and liver that transdifferentiate to a myofibroblastic state in the context of tissue injury. Beyond having roles in tissue homeostasis, stellate cells are increasingly implicated in pathological fibrogenic and inflammatory programs that contribute to tissue fibrosis and that constitute a growth-permissive tumor microenvironment. Although the capacity of stellate cells for extracellular matrix production and remodeling has long been appreciated, recent research efforts have demonstrated diverse roles for stellate cells in regulation of epithelial cell fate, immune modulation, and tissue health. Our present understanding of stellate cell biology in health and disease is discussed here, as are emerging means to target these multifaceted cells for therapeutic benefit.


Subject(s)
Hepatic Stellate Cells/metabolism , Inflammation/genetics , Neoplasms/genetics , Pancreatic Stellate Cells/metabolism , Cell Transdifferentiation/genetics , Hepatic Stellate Cells/pathology , Humans , Inflammation/pathology , Liver/metabolism , Liver/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Neoplasms/pathology , Pancreas/injuries , Pancreas/metabolism , Pancreas/pathology , Pancreatic Stellate Cells/pathology , Tumor Microenvironment/genetics , Wound Healing
20.
Cell ; 165(3): 668-78, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27062926

ABSTRACT

A key feature of inflammation is the timely recruitment of leukocytes, including monocytes, from blood into tissues, the latter maturing into macrophages over a period of 2-3 days. Using multi-channel spinning disk microscopy, we identified a rapid pathway of macrophage recruitment into an injured organ via a non-vascular route requiring no maturation from monocytes. In response to a sterile injury in liver, a reservoir of fully mature F4/80(hi)GATA6(+) peritoneal cavity macrophages rapidly invaded into afflicted tissue via direct recruitment across the mesothelium. The invasion was dependent on CD44 and DAMP molecule ATP and resulted in rapid replication and switching of macrophage toward an alternatively activated phenotype. These macrophages dismantled the nuclei of necrotic cells releasing DNA and forming a cover across the injury site. Rapid invasion of mature macrophages from body cavity with capacity for induction of reparative phenotype may impact altered tissues ranging from trauma to infections to cancer. VIDEO ABSTRACT.


Subject(s)
Cell Movement , Liver/physiology , Macrophages, Peritoneal/cytology , Wound Healing , Animals , Disease Models, Animal , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Inflammation , Leukocytes/immunology , Leukocytes/metabolism , Liver/injuries , Liver/pathology , Mice , Mice, Knockout , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL