Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 659
Filter
1.
Immunol Cell Biol ; 102(5): 381-395, 2024.
Article in English | MEDLINE | ID: mdl-38629182

ABSTRACT

Resident macrophages of various mammalian organs are characterized by several distinctive features in their gene expression profile and phenotype, including involvement in the regulation of organ functions, as well as reduced sensitivity to proinflammatory activation factors. The reasons for the formation of such a specific phenotype remain the subject of intensive research. Some papers emphasize the role of the origin of organ macrophages. Other studies indicate that monocytes that develop in the red bone marrow are also able to form resident macrophages with a phenotype characteristic of a particular organ, but this requires appropriate microenvironmental conditions. In this article, we studied the possibility of differentiation of monocyte-derived macrophages into cells with a Kupffer-like phenotype under the influence of the main stromal components of Kupffer cells macrophage niche: Ito cells, liver sinusoid endotheliocytes and hepatocyte growth factor (HGF). It was found that Kupffer cells are characterized by several features, including increased expression of transcription factors Spic and Id3, as well as MUP family genes, Clusterin and Ngp genes. In addition, Kupffer cells were characterized by a higher proliferative activity. The expression of marker genes of Kupffer cells (i.e. Id3, Spic, Marco and Timd4) increased in monocyte-derived macrophages during coculture with Ito cells, liver sinusoid endothelial cells, macrophage colony-stimulating factor and HGF cells only by 3 days. However, the expression level of these genes was always higher in Kupffer cells. In addition, a complete coincidence of the expressed gene profile in monocyte-derived macrophages and Kupffer cells did not occur even after 3 days of culturing.


Subject(s)
Cell Differentiation , Cellular Microenvironment , Kupffer Cells , Macrophages , Phenotype , Kupffer Cells/metabolism , Kupffer Cells/cytology , Macrophages/metabolism , Animals , Monocytes/metabolism , Monocytes/cytology , Hepatocyte Growth Factor/metabolism , Endothelial Cells/metabolism , Coculture Techniques , Humans , Cell Proliferation , Cells, Cultured , Liver/cytology , Liver/metabolism , Mice
2.
Nature ; 626(8000): 864-873, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38326607

ABSTRACT

Macrophage activation is controlled by a balance between activating and inhibitory receptors1-7, which protect normal tissues from excessive damage during infection8,9 but promote tumour growth and metastasis in cancer7,10. Here we report that the Kupffer cell lineage-determining factor ID3 controls this balance and selectively endows Kupffer cells with the ability to phagocytose live tumour cells and orchestrate the recruitment, proliferation and activation of natural killer and CD8 T lymphoid effector cells in the liver to restrict the growth of a variety of tumours. ID3 shifts the macrophage inhibitory/activating receptor balance to promote the phagocytic and lymphoid response, at least in part by buffering the binding of the transcription factors ELK1 and E2A at the SIRPA locus. Furthermore, loss- and gain-of-function experiments demonstrate that ID3 is sufficient to confer this potent anti-tumour activity to mouse bone-marrow-derived macrophages and human induced pluripotent stem-cell-derived macrophages. Expression of ID3 is therefore necessary and sufficient to endow macrophages with the ability to form an efficient anti-tumour niche, which could be harnessed for cell therapy in cancer.


Subject(s)
Inhibitor of Differentiation Proteins , Kupffer Cells , Neoplasms , Animals , Humans , Mice , Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage , Induced Pluripotent Stem Cells/cytology , Inhibitor of Differentiation Proteins/deficiency , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Kupffer Cells/cytology , Kupffer Cells/immunology , Kupffer Cells/metabolism , Liver/immunology , Liver/pathology , Macrophage Activation , Neoplasm Proteins , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phagocytosis
3.
Nat Commun ; 12(1): 7046, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857782

ABSTRACT

Reconstruction of heterogeneity through single cell transcriptional profiling has greatly advanced our understanding of the spatial liver transcriptome in recent years. However, global transcriptional differences across lobular units remain elusive in physical space. Here, we apply Spatial Transcriptomics to perform transcriptomic analysis across sectioned liver tissue. We confirm that the heterogeneity in this complex tissue is predominantly determined by lobular zonation. By introducing novel computational approaches, we enable transcriptional gradient measurements between tissue structures, including several lobules in a variety of orientations. Further, our data suggests the presence of previously transcriptionally uncharacterized structures within liver tissue, contributing to the overall spatial heterogeneity of the organ. This study demonstrates how comprehensive spatial transcriptomic technologies can be used to delineate extensive spatial gene expression patterns in the liver, indicating its future impact for studies of liver function, development and regeneration as well as its potential in pre-clinical and clinical pathology.


Subject(s)
Genetic Heterogeneity , Liver/metabolism , Transcriptome , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Erythroblasts/cytology , Erythroblasts/metabolism , Female , Gene Expression Profiling , Gene Ontology , Hepatocytes/cytology , Hepatocytes/metabolism , Kupffer Cells/cytology , Kupffer Cells/metabolism , Liver/cytology , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Annotation , Neutrophils/cytology , Neutrophils/metabolism
4.
Arq. bras. med. vet. zootec. (Online) ; 73(4): 868-876, Jul.-Aug. 2021. graf, mapas, ilus
Article in English | LILACS, VETINDEX | ID: biblio-1285271

ABSTRACT

The melanomacrophage centers (MMCs) in the liver of fish are indicators of environmental conditions, as they are involved in xenobiotic biotransformation. The objective of this study was to evaluate the number of MMC in the liver of juveniles and adults of Sciades herzbergii from areas with different levels of contamination. The fish were caught at three points (reference - A1, potentially impacted - A2 and contaminated - A3), in São José bay (Maranhão, Brazil), in four samples. The livers were subjected to the standard histological procedure and 5µm sections were stained with hematoxylin-eosin. In livers of A2 adult individuals (260.50±161.50 MMCs / mm²) they presented a greater number of MMCs when compared to A3 adults (60.00 ± 30.10 MMCs / mm²). Juveniles showed considerable values in A1 (100.00 ± 0.00 MMCs/mm²) and A2 (95.33 ± 33.00 MMCs / mm²) compared to juveniles in A3 (49.00±0.00 MMCs/mm²). These high values are unexpected for young people. The average number of MMC correlated with the rainy season in the region. The use of hepatic MMCs as a biomarker of exposure to pollutants, in particular substances from fisheries systems, such as ammonia and nitrite, proved to be adequate to differentiate areas with different levels of impacts.(AU)


Os centros melanomacrófagos (MMCs) no fígado de peixes são indicadores das condições ambientais, pois estão envolvidos na biotransformação xenobiótica. O objetivo deste estudo foi avaliar o número de MMC no fígado de juvenis e adultos de Sciades herzbergii de áreas com diferentes níveis de contaminação. Os peixes foram capturados em três pontos (referência - A1; potencialmente impactado - A2; e contaminado - A3), na baía de São José (Maranhão, Brasil), em quatro amostras. Os fígados foram submetidos ao procedimento histológico padrão e cortes de 5µm foram corados com hematoxilina-eosina. Em fígados de indivíduos adultos A2 (260,50±161,50 MMCs/mm²), eles apresentaram maior número de MMCs quando comparados aos adultos A3 (60,00±30,10 MMCs/mm²). Os juvenis apresentaram valores elevados em A1 (100,00 ± 0,00 MMCs/mm²) e A2 (95,33±33,00 MMCs/mm²) quando comparados aos juvenis em A3 (49,00±0,00 MMCs/mm²). Esses altos valores são inesperados para os jovens. O número médio de MMC correlacionou-se com a época chuvosa na região. A utilização de MMCs hepáticos como biomarcador de exposição a poluentes, em particular substâncias provenientes de sistemas pesqueiros, como amônia e nitrito, mostrou-se adequada para diferenciar áreas com diferentes níveis de impactos.(AU)


Subject(s)
Animals , Catfishes , Environmental Biomarkers , Biological Monitoring/methods , Kupffer Cells , Kupffer Cells/cytology , Environmental Pollution/analysis
5.
Eur J Immunol ; 51(10): 2452-2463, 2021 10.
Article in English | MEDLINE | ID: mdl-34324208

ABSTRACT

Abundant long-lived liver-resident macrophages, termed Kupffer cells, are activated during chronic liver injury. They secrete both pro-inflammatory and pro-fibrotic cytokines, which act on hepatic stellate cells causing their transdifferentiation into myofibroblasts that deposit collagen. In other tissues, wound-associated macrophages go further, and transdifferentiate into fibrocytes, secreting collagen themselves. We tested Kupffer cells for this property in two experimental models: mixed non-parenchymal cell culture, and precision-cut liver slice culture. Using the Emr1-Cre transgene as a driver and the RiboTag transgene as a reporter, we found that Kupffer cells undergo transdifferentiation under these circumstances. Over time, they lose the expression of both Kupffer cell-specific and macrophage-specific genes and the transcription factors that control their expression, and they begin to express multiple genes and proteins characteristic of either myofibroblasts or tissue fibroblasts. These effects were strongly conserved between non-parenchymal cell culture and liver tissue slice culture, arguing that such transdifferentiation is a conserved function of Kupffer cells. We conclude that in addition to supporting fibrosis through an action on stellate cells, Kupffer cells also participate in liver fibrosis through transdifferentiation into fibrocytes.


Subject(s)
Biomarkers , Cell Transdifferentiation , Kupffer Cells/cytology , Kupffer Cells/metabolism , Signal Transduction , Animals , Cell Transdifferentiation/genetics , Cells, Cultured , Fibrosis/genetics , Fibrosis/metabolism , Gene Expression Regulation/drug effects , Immunohistochemistry , Mice , Phenotype , Transcription Factors/genetics
6.
Int J Mol Sci ; 22(9)2021 Apr 22.
Article in English | MEDLINE | ID: mdl-33922101

ABSTRACT

3D cell culture systems are widely used to study disease mechanisms and therapeutic interventions. Multicellular liver microtissues (MTs) comprising HepaRG, hTERT-HSC and THP-1 maintain multicellular interactions and physiological properties required to mimic liver fibrosis. However, the inherent complexity of multicellular 3D-systems often hinders the discrimination of cell type specific responses. Here, we aimed at applying single cell sequencing (scRNA-seq) to discern the molecular responses of cells involved in the development of fibrosis elicited by TGF-ß1. To obtain single cell suspensions from the MTs, an enzymatic dissociation method was optimized. Isolated cells showed good viability, could be re-plated and cultured in 2D, and expressed specific markers determined by scRNA-seq, qRT-PCR, ELISA and immunostaining. The three cell populations were successfully clustered using supervised and unsupervised methods based on scRNA-seq data. TGF-ß1 led to a fibrotic phenotype in the MTs, detected as decreased albumin and increased αSMA expression. Cell-type specific responses to the treatment were identified for each of the three cell types. They included HepaRG damage characterized by a decrease in cellular metabolism, prototypical inflammatory responses in THP-1s and extracellular matrix remodeling in hTERT-HSCs. Furthermore, we identified novel cell-specific putative fibrosis markers in hTERT-HSC (COL15A1), and THP-1 (ALOX5AP and LAPTM5).


Subject(s)
Biomarkers/metabolism , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Kupffer Cells/metabolism , Liver Cirrhosis/metabolism , Single-Cell Analysis/methods , Transforming Growth Factor beta1/pharmacology , Cell Culture Techniques , Cell Proliferation , Gene Expression Regulation , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Kupffer Cells/cytology , Kupffer Cells/drug effects , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Prognosis
7.
Cell Prolif ; 54(4): e13022, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33686740

ABSTRACT

OBJECTIVES: This study aimed to investigate the protective effect of SCARF1 on acute rejection (AR), phagocytic clearance of Kupffer cells (KCs), M2 polarization and the exact mechanism underlying these processes. METHODS: AAV was transfected into the portal vein of rats, and AR and immune tolerance (IT) models of liver transplantation were established. Liver tissue and blood samples were collected. The level of SCARF1 was detected via WB and immunohistochemical staining. Pathological changes in liver tissue were detected using HE staining. Apoptotic cells were detected using TUNEL staining. KC polarization was assessed via immunohistochemical staining. Primary KCs were isolated and co-cultured with apoptotic T lymphocytes. Phagocytosis of apoptotic cells and polarization of KCs were both detected using immunofluorescence. Calcium concentration was determined using immunofluorescence and a fluorescence microplate reader. The levels of PI3K, p-AKT and P-STAT3 were assessed via WB and immunofluorescence. RESULTS: Compared to the IT group, the level of SCARF1 was significantly decreased in the AR group. Overexpression of SCARF1 in KCs improved AR and liver function markers. Enhanced phagocytosis mediated by SCARF1 is beneficial for improving the apoptotic clearance of AR and promoting M2 polarization of KCs. SCARF1-mediated enhancement of phagocytosis promotes increased calcium concentration in KCs, thus further activating the PI3K-AKT-STAT3 signalling pathway. CONCLUSIONS: SCARF1 promotes the M2 polarization of KCs by promoting phagocytosis through the calcium-dependent PI3K-AKT-STAT3 signalling pathway.


Subject(s)
Calcium/metabolism , Liver Transplantation , Scavenger Receptors, Class F/metabolism , Signal Transduction , Animals , Apoptosis , Cell Polarity , Cells, Cultured , Coculture Techniques , Kupffer Cells/cytology , Kupffer Cells/metabolism , Liver/metabolism , Liver/pathology , Male , Phagocytosis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred Lew , STAT3 Transcription Factor/metabolism , Scavenger Receptors, Class F/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
8.
Hepatol Commun ; 5(2): 217-233, 2021 02.
Article in English | MEDLINE | ID: mdl-33553970

ABSTRACT

Nonalcoholic steatohepatitis (NASH), an advanced stage of nonalcoholic fatty liver disease (NAFLD), is a rapidly growing and global health problem compounded by the current absence of specific treatments. A major limiting factor in the development of new NASH therapies is the absence of models that capture the unique cellular structure of the liver microenvironment and recapitulate the complexities of NAFLD progression to NASH. Organ-on-a-chip platforms have emerged as a powerful approach to dynamically model diseases and test drugs. Herein, we describe a NASH-on-a-chip platform. Four main types of human primary liver cells (hepatocytes [HCs], Kupffer cells, liver sinusoidal endothelial cells, and hepatic stellate cells [HSCs]) were cocultured under microfluidic dynamics. Our chip-based model successfully recapitulated a functional liver cellular microenvironment with stable albumin and urea secretion for at least 2 weeks. Exposing liver chips to a lipotoxic environment led to gradual development of NASH phenotypic characteristics, including intracellular lipid accumulation, hepatocellular ballooning, HSC activation, and elevation of inflammatory and profibrotic markers. Further, exposure of the chip to elafibranor, a drug under study for the therapy of NASH, inhibited the development of NASH-specific hallmarks, causing an ~8-fold decrease in intracellular lipids, a 3-fold reduction in number of ballooned HCs, a significant reduction in HSC activation, and a significant decrease in the levels of inflammatory and profibrotic markers compared with controls. Conclusion: We have successfully developed a microfluidic NASH-on-a-chip platform that recapitulates the main NASH histologic endpoints in a single chip and that can emerge as a powerful noninvasive, human-relevant, in vitro platform to study disease pathogenesis and develop novel anti-NASH drugs.


Subject(s)
Coculture Techniques , Lab-On-A-Chip Devices , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/physiopathology , Chalcones/pharmacology , Endothelial Cells/cytology , Hepatic Stellate Cells/cytology , Hepatocytes/cytology , Humans , Inflammation , Kupffer Cells/cytology , Liver , Propionates/pharmacology
9.
Biochim Biophys Acta Gen Subj ; 1865(4): 129559, 2021 04.
Article in English | MEDLINE | ID: mdl-32084396

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs) are a diverse group of membrane-bound nanovesicles potentially released by every cell. With the liver's unique ensemble of cells and its fundamental physiological tasks, elucidating the role of EV-mediated hepatic cellular crosstalk and their role in different pathologies has been gaining the attention of many scientists. SCOPE OF REVIEW: The present review shifts the perspective into practice: we aim to critically discuss the methods used to purify and to biochemically analyse EVs from specific liver resident cells, including hepatocytes, hepatic stellate cells, cholangiocytes, liver sinusoidal endothelial cells, Kupffer cells, liver stem cells. The review offers a reference guide to current approaches. MAJOR CONCLUSIONS: Strategies for EV isolation and characterization are as varied as the research groups performing them. We present main advantages and disadvantages for the methods, highlighting common causes for concern, such as FBS handling, reporting of cell viability, EV yield and storage, differences in differential centrifugations, suboptimal method descriptions, and method transferability. We both looked at how adaptable the research between human and rodent cells in vitro is, and also assessed how well either of them translates to ex vivo settings. GENERAL SIGNIFICANCE: We reviewed methodological practices for the isolation and analysis of liver-derived EVs, making a cell type specific user guide that shows where to start, what has worked so far and to what extent. We critically discussed room for improvement, placing a particular focus on working towards a potential standardization of methods.


Subject(s)
Extracellular Vesicles/chemistry , Liver/cytology , Animals , Centrifugation/methods , Endothelial Cells/chemistry , Endothelial Cells/cytology , Hepatic Stellate Cells/chemistry , Hepatic Stellate Cells/cytology , Hepatocytes/chemistry , Hepatocytes/cytology , Humans , Kupffer Cells/chemistry , Kupffer Cells/cytology , Liver/chemistry , Stem Cells/chemistry , Stem Cells/cytology
10.
Nature ; 589(7840): 131-136, 2021 01.
Article in English | MEDLINE | ID: mdl-33239787

ABSTRACT

The liver connects the intestinal portal vasculature with the general circulation, using a diverse array of immune cells to protect from pathogens that translocate from the gut1. In liver lobules, blood flows from portal triads that are situated in periportal lobular regions to the central vein via a polarized sinusoidal network. Despite this asymmetry, resident immune cells in the liver are considered to be broadly dispersed across the lobule. This differs from lymphoid organs, in which immune cells adopt spatially biased positions to promote effective host defence2,3. Here we used quantitative multiplex imaging, genetic perturbations, transcriptomics, infection-based assays and mathematical modelling to reassess the relationship between the localization of immune cells in the liver and host protection. We found that myeloid and lymphoid resident immune cells concentrate around periportal regions. This asymmetric localization was not developmentally controlled, but resulted from sustained MYD88-dependent signalling induced by commensal bacteria in liver sinusoidal endothelial cells, which in turn regulated the composition of the pericellular matrix involved in the formation of chemokine gradients. In vivo experiments and modelling showed that this immune spatial polarization was more efficient than a uniform distribution in protecting against systemic bacterial dissemination. Together, these data reveal that liver sinusoidal endothelial cells sense the microbiome, actively orchestrating the localization of immune cells, to optimize host defence.


Subject(s)
Gastrointestinal Microbiome/immunology , Liver/immunology , Liver/microbiology , Symbiosis/immunology , Animals , Bacteria/immunology , Bacteria/isolation & purification , Cell Separation , Chemokine CXCL9/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Female , Humans , Kupffer Cells/cytology , Kupffer Cells/immunology , Kupffer Cells/metabolism , Liver/blood supply , Liver/cytology , Lymphocytes/immunology , Male , Mice , Models, Immunological , Molecular Imaging , Myeloid Cells/immunology , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Symbiosis/genetics , Transcriptome
11.
Bull Exp Biol Med ; 170(1): 40-45, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33222081

ABSTRACT

Hepatocyte nuclear factor (HNF-6) is a liver-specific protein and a key component in the differentiation process during the development of mature liver. The immunohistochemical staining and RT-PCR techniques were employed to examine the expression of HNF-6 and proliferation of Ki-67+ cells during the early regeneration of the liver on postsurgery in 3, 6, 12, and 24 h in original model of partial hepatectomy in rats. The earliest proliferating (Ki-67+) cells were observed in 3 h after surgery in liver sinusoids (liver macrophages) and then in liver parenchyma. Expression of HNF-6 in hepatocytes and epithelial cells of the bile ducts attained maximum in 6 h after surgery. At later terms, this parameter somewhat decreased, but still surpassed the control level.


Subject(s)
Hepatocyte Nuclear Factor 6/genetics , Hepatocytes/metabolism , Kupffer Cells/metabolism , Liver Regeneration/genetics , Liver/metabolism , Animals , Bile Ducts/metabolism , Bile Ducts/surgery , Cell Proliferation , Female , Gene Expression Regulation , Hepatectomy/methods , Hepatocyte Nuclear Factor 6/metabolism , Hepatocytes/cytology , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Kupffer Cells/cytology , Liver/surgery , Male , Rats , Rats, Sprague-Dawley
12.
Front Immunol ; 11: 2108, 2020.
Article in English | MEDLINE | ID: mdl-33101269

ABSTRACT

Human liver myeloid cells are imperfectly defined, but it is broadly agreed that cells of stellate appearance in situ, expressing the markers CD11b and CD68, are the liver's resident macrophages, classically termed Kupffer cells. Recent investigations using single cell RNA sequencing and unsupervised clustering algorithms suggest there are two populations of cells with the characteristics of tissue macrophages in human liver. We therefore analyzed dissociated human liver tissue using the markers CD11b and CD68 to define macrophage-like cells and found within this population two subsets that differ in their expression of multiple surface markers. These subsets were FACS-sorted based on CD32 expression, and gene expression analysis identified them with human liver myeloid cell subsets that were previously defined by two independent single cell RNA sequencing studies. Using qRT-PCR we found that the two subsets differed in the expression of genes associated with T cell activation and immunosuppression, suggesting distinct roles in T cell tolerance. In addition, one subset expressed two markers, CD1C and CD11c, more often seen on classical dendritic cells. Criteria used to distinguish macrophages from dendritic cells in other tissues may need to be revised in the human liver.


Subject(s)
Antigens, CD1/immunology , CD11 Antigens/immunology , Glycoproteins/immunology , Integrin alpha Chains/immunology , Kupffer Cells/immunology , Liver/immunology , Receptors, IgG/immunology , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , CD11b Antigen/immunology , Flow Cytometry , Gene Expression Regulation/immunology , Humans , Kupffer Cells/cytology , Liver/cytology
13.
Immunity ; 53(3): 641-657.e14, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32888418

ABSTRACT

Metabolic-associated fatty liver disease (MAFLD) represents a spectrum of disease states ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). Hepatic macrophages, specifically Kupffer cells (KCs), are suggested to play important roles in the pathogenesis of MAFLD through their activation, although the exact roles played by these cells remain unclear. Here, we demonstrated that KCs were reduced in MAFLD being replaced by macrophages originating from the bone marrow. Recruited macrophages existed in two subsets with distinct activation states, either closely resembling homeostatic KCs or lipid-associated macrophages (LAMs) from obese adipose tissue. Hepatic LAMs expressed Osteopontin, a biomarker for patients with NASH, linked with the development of fibrosis. Fitting with this, LAMs were found in regions of the liver with reduced numbers of KCs, characterized by increased Desmin expression. Together, our data highlight considerable heterogeneity within the macrophage pool and suggest a need for more specific macrophage targeting strategies in MAFLD.


Subject(s)
Bone Marrow Cells/cytology , Macrophage Activation/immunology , Macrophages/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Osteopontin/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Desmin/metabolism , Female , Kupffer Cells/cytology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Proteome/metabolism , Transcriptome/genetics
14.
Methods Mol Biol ; 2164: 1-10, 2020.
Article in English | MEDLINE | ID: mdl-32607877

ABSTRACT

Kupffer cells and hepatocytes maintain liver homeostasis. These cells could be separated based on their size and weight, by centrifugation using a density gradient after a liver perfusion. Here, we describe a methodology to isolate both Kupffer cells and hepatocytes from a single mouse, which provides the unique advantage of studying these two cell types from the same liver.


Subject(s)
Centrifugation/methods , Hepatocytes/cytology , Kupffer Cells/cytology , Liver/cytology , Animals , Cell Separation/methods , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Perfusion/methods
15.
Methods Mol Biol ; 2164: 11-13, 2020.
Article in English | MEDLINE | ID: mdl-32607878

ABSTRACT

Liver macrophages (LMs) are phagocytic cells that play an important role in many liver disorders due to their ability to respond to a variety of stimuli and activating signals.It is currently debated whether LMs activation from an anti-inflammatory to a proinflammatory phenotype contributes to obesity-induced metabolic diseases. We recently found that LMs can produce noninflammatory factors, such as the protein IGFBP7, able to directly regulate hepatic glucose production and lipid accumulation in the liver. However, while in a mouse model of obesity and insulin resistance LM-Igfbp7 expression is pathologically increased, in obese insulin-resistant patients LM-IGFBP7 is edited at RNA level independently of an effect on its expression. This discrepancy between results in animals and humans confirms the importance to perform molecular investigation directly on human's isolated cells. Here, we describe a protocol to isolate liver macrophages from human liver biopsy .


Subject(s)
Kupffer Cells/cytology , Liver/cytology , Animals , Biopsy/methods , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Resistance/physiology , Insulin-Like Growth Factor Binding Proteins/metabolism , Kupffer Cells/metabolism , Lipid Metabolism/physiology , Liver/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Obesity/metabolism , Obesity/pathology , RNA/metabolism
16.
Methods Mol Biol ; 2164: 45-53, 2020.
Article in English | MEDLINE | ID: mdl-32607882

ABSTRACT

Kupffer cells are the liver-resident macrophages and represent the first line of defense between the pathogens circulating from the intestines through the portal vein and systemic circulation. Recent works have highlighted the complex heterogeneity of macrophage functions and origins, thus raising awareness on the need for a better characterization of macrophage populations. The immunohistochemistry method here described, allows for a rapid distinction between Kupffer cells and monocyte-derived macrophages present on formalin-fixed, paraffin-embedded mouse liver samples. This protocol has been optimized for its reproducibility, reliability, and simplicity.


Subject(s)
Fluorescent Antibody Technique/methods , Formaldehyde/chemistry , Kupffer Cells/cytology , Macrophages/cytology , Paraffin Embedding/methods , Tissue Fixation/methods , Animals , Immunohistochemistry/methods , Liver/cytology , Mice , Reproducibility of Results
17.
Methods Mol Biol ; 2164: 87-99, 2020.
Article in English | MEDLINE | ID: mdl-32607886

ABSTRACT

Kupffer cells are the liver-resident macrophages lining the sinusoids and are mostly known for their role of scavengers, as crucial keepers of organ integrity. But due to the many fundamental functions of the liver notably linked to detoxication, metabolism, protein synthesis, or immunology, Kupffer cells are exposed to a dynamic environment and constantly adapt themselves by modulating their gene and protein expressions. In this context, the characterization of these cells at steady-state and upon challenges may be limited by the classical microscopy or flow cytometry which allow for the use of only few selected markers. On the other end, transcriptomic approach, although being very powerful, can be costly and time-consuming. So mass cytometry offers a good compromise, allowing for the monitoring of a representative set of markers (up to 40) in a simple experiment. Herein, we describe a straightforward experimental and analysis workflow for Kupffer cell characterization by mass cytometry.


Subject(s)
Flow Cytometry/methods , Kupffer Cells/cytology , Animals , Liver/cytology , Macrophages/cytology , Mice
18.
PLoS One ; 15(6): e0234038, 2020.
Article in English | MEDLINE | ID: mdl-32492075

ABSTRACT

Extracellular adenosine triphosphate (eATP) released by damaged cells, and its purinergic receptors, comprise a crucial signaling network after injury. Purinergic receptor P2X7 (P2RX7), a major driver of NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and IL-1ß processing, has been shown to play a role in liver injury in murine diet- and chemically-induced liver injury models. It is unclear, however, whether P2RX7 plays a role in non-alcoholic steatohepatitis (NASH) and which cell type is the main target of P2RX7 pharmacological inhibition. Here, we report that P2RX7 is expressed by infiltrating monocytes and resident Kupffer cells in livers from NASH-affected individuals. Using primary isolated human cells, we demonstrate that P2RX7 expression in CD14+ monocytes and Kupffer cells primarily mediates IL-1ß release. In addition, we show that pharmacological inhibition of P2RX7 in monocytes and Kupffer cells, blocks IL-1ß release, reducing hepatocyte caspase 3/7 activity, IL-1ß-mediated CCL2 and CCL5 chemokine gene expression and secretion, and hepatic stellate cell (HSC) procollagen secretion. Consequently, in a chemically-induced nonhuman primate model of liver fibrosis, treatment with a P2RX7 inhibitor improved histological characteristics of NASH, protecting from liver inflammation and fibrosis. Taken together, these findings underscore the critical role of P2RX7 in the pathogenesis of NASH and implicate P2RX7 as a promising therapeutic target for the management of this disease.


Subject(s)
Inflammation/prevention & control , Liver Cirrhosis/drug therapy , Purinergic P2X Receptor Antagonists/therapeutic use , Receptors, Purinergic P2X7/metabolism , Animals , Caspase 3/metabolism , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inflammation/pathology , Interleukin-1beta/metabolism , Kupffer Cells/cytology , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Lipopolysaccharides/pharmacology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Macaca fascicularis , Male , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Procollagen/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/genetics
19.
Bull Exp Biol Med ; 168(4): 556-560, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32157510

ABSTRACT

Differences in the gene expression profiles in resident macrophages (in particular, Kupffer cells) and monocytes were revealed. However, these differences in gene expression profiles do not allow considering resident liver macrophages as purely M2 macrophages and monocytes as purely M1 macrophages. At the same time, a significant number of the genes upregulated in Kupffer cells are associated with normal regulation of liver functions (Arg 1, Flt, iNOs, and Kng). In monocytes, the expression of genes Alox15, Alox12, Tlr2, Tlr4, Tlr7, and Tlr8 (typical functional genes of macrophages) was also upregulated in comparison with Kupffer cells.


Subject(s)
Cell Lineage/genetics , Kupffer Cells/metabolism , Liver/metabolism , Monocytes/metabolism , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Arginase/genetics , Arginase/metabolism , Biomarkers/metabolism , Gene Expression , Immunophenotyping , Kupffer Cells/classification , Kupffer Cells/cytology , Liver/cytology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Monocytes/classification , Monocytes/cytology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
20.
Nat Commun ; 11(1): 1269, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152267

ABSTRACT

Multicellular rosettes are transient epithelial structures that serve as intermediates during diverse organ formation. We have identified a unique contributor to rosette formation in zebrafish Kupffer's vesicle (KV) that requires cell division, specifically the final stage of mitosis termed abscission. KV utilizes a rosette as a prerequisite before forming a lumen surrounded by ciliated epithelial cells. Our studies identify that KV-destined cells remain interconnected by cytokinetic bridges that position at the rosette's center. These bridges act as a landmark for directed Rab11 vesicle motility to deliver an essential cargo for lumen formation, CFTR (cystic fibrosis transmembrane conductance regulator). Here we report that premature bridge cleavage through laser ablation or inhibiting abscission using optogenetic clustering of Rab11 result in disrupted lumen formation. We present a model in which KV mitotic cells strategically place their cytokinetic bridges at the rosette center, where Rab11-associated vesicles transport CFTR to aid in lumen establishment.


Subject(s)
Cell Division/physiology , Cell Polarity/physiology , Embryo, Nonmammalian/physiology , Embryonic Development/physiology , Kupffer Cells/physiology , Organogenesis/physiology , Zebrafish/embryology , Animals , Cell Line , Cell Movement , Cilia/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Kupffer Cells/cytology , Mitosis , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...