Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Nat Immunol ; 25(1): 155-165, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38102487

ABSTRACT

In mouse peritoneal and other serous cavities, the transcription factor GATA6 drives the identity of the major cavity resident population of macrophages, with a smaller subset of cavity-resident macrophages dependent on the transcription factor IRF4. Here we showed that GATA6+ macrophages in the human peritoneum were rare, regardless of age. Instead, more human peritoneal macrophages aligned with mouse CD206+ LYVE1+ cavity macrophages that represent a differentiation stage just preceding expression of GATA6. A low abundance of CD206+ macrophages was retained in C57BL/6J mice fed a high-fat diet and in wild-captured mice, suggesting that differences between serous cavity-resident macrophages in humans and mice were not environmental. IRF4-dependent mouse serous cavity macrophages aligned closely with human CD1c+CD14+CD64+ peritoneal cells, which, in turn, resembled human peritoneal CD1c+CD14-CD64- cDC2. Thus, major populations of serous cavity-resident mononuclear phagocytes in humans and mice shared common features, but the proportions of different macrophage differentiation stages greatly differ between the two species, and dendritic cell (DC2)-like cells were especially prominent in humans.


Subject(s)
Macrophages, Peritoneal , Macrophages , Humans , Mice , Animals , Mice, Inbred C57BL , Macrophages/metabolism , Macrophages, Peritoneal/metabolism , Cell Differentiation , Dendritic Cells
2.
Proc Natl Acad Sci U S A ; 109(11): E630-9, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22247292

ABSTRACT

The dicistrovirus intergenic region internal ribosome entry site (IRES) utilizes a unique mechanism, involving P-site tRNA mimicry, to directly assemble 80S ribosomes and initiate translation at a specific non-AUG codon in the ribosomal A site. A subgroup of dicistrovirus genomes contains an additional stem-loop 5'-adjacent to the IRES and a short open reading frame (ORFx) that overlaps the viral structural polyprotein ORF (ORF2) in the +1 reading frame. Using mass spectrometry and extensive mutagenesis, we show that, besides directing ORF2 translation, the Israeli acute paralysis dicistrovirus IRES also directs ORFx translation. The latter is mediated by a UG base pair adjacent to the P-site tRNA-mimicking domain. An ORFx peptide was detected in virus-infected honey bees by multiple reaction monitoring mass spectrometry. Finally, the 5' stem-loop increases IRES activity and may couple translation of the two major ORFs of the virus. This study reveals a novel viral strategy in which a tRNA-like IRES directs precise, initiator Met-tRNA-independent translation of two overlapping ORFs.


Subject(s)
Nucleic Acid Conformation , RNA, Transfer/chemistry , RNA, Transfer/genetics , Reading Frames/genetics , Ribosomes/genetics , Selection, Genetic , Animals , Base Pairing/genetics , Base Sequence , Bees/virology , DNA, Complementary/genetics , DNA, Intergenic/genetics , Dicistroviridae/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Molecular Sequence Data , Peptide Chain Initiation, Translational/genetics , Pupa/virology
3.
Biochemistry ; 53(5): 932-46, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24447242

ABSTRACT

Ryanodine receptors (RyRs) are large ion channels that are responsible for the release of Ca(2+) from the sarcoplasmic/endoplasmic reticulum. Calmodulin (CaM) is a Ca(2+) binding protein that can affect the channel open probability at both high and low Ca(2+) concentrations, shifting the Ca(2+) dependencies of channel opening in an isoform-specific manner. Here we analyze the binding of CaM and its individual domains to three different RyR regions using isothermal titration calorimetry. We compared binding to skeletal muscle (RyR1) and cardiac (RyR2) isoforms, under both Ca(2+)-loaded and Ca(2+)-free conditions. CaM can bind all three regions in both isoforms, but the binding modes differ appreciably in two segments. The results highlight a Ca(2+)/CaM and apoCaM binding site in the C-terminal fifth of the channel. This binding site is the target for malignant hyperthermia and central core disease mutations in RyR1, which affect the energetics and mode of CaM binding.


Subject(s)
Calmodulin/chemistry , Ryanodine Receptor Calcium Release Channel/chemistry , Animals , Binding Sites , Calmodulin/genetics , Humans , Malignant Hyperthermia/genetics , Muscle, Skeletal/metabolism , Mutation , Myocardium/metabolism , Myopathy, Central Core/genetics , Protein Binding , Protein Isoforms/chemistry , Rabbits , Rats , Thermodynamics
4.
bioRxiv ; 2023 Aug 25.
Article in English | MEDLINE | ID: mdl-36824926

ABSTRACT

Hepatic stellate cells (HSC) are non-parenchymal liver cells that produce extracellular matrix comprising fibrotic lesions in chronic liver diseases. Prior work demonstrated that mitochondrial pyruvate carrier (MPC) inhibitors suppress HSC activation and fibrosis in a mouse model of metabolic dysfunction-associated steatohepatitis (MASH). In the present study, pharmacologic or genetic inhibition of the MPC in HSC decreased expression of markers of activation in vitro. MPC knockdown also reduced the abundance of several intermediates of the TCA cycle, and diminished α-ketoglutarate played a key role in attenuating HSC activation by suppressing hypoxia inducible factor-1α signaling. On high fat diets, mice with HSC-specific MPC deletion exhibited reduced circulating transaminases, numbers of HSC, and hepatic expression of markers of HSC activation and inflammation compared to wild-type mice. These data suggest that MPC inhibition modulates HSC metabolism to attenuate activation and illuminate mechanisms by which MPC inhibitors could prove therapeutically beneficial for treating MASH.

5.
JHEP Rep ; 5(11): 100877, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37869071

ABSTRACT

Background & Aims: Metabolic dysfunction-associated fatty liver disease (MAFLD) is a common complication of obesity with a hallmark feature of hepatic steatosis. Recent data from animal models of MAFLD have demonstrated substantial changes in macrophage composition in the fatty liver. In humans, the relationship between liver macrophage heterogeneity and liver steatosis is less clear. Methods: Liver tissue from 21 participants was collected at time of bariatric surgery and analysed using flow cytometry, immunofluorescence, and H&E microscopy. Single-cell RNA sequencing was also conducted on a subset of samples (n = 3). Intrahepatic triglyceride content was assessed via MRI and tissue histology. Mouse models of hepatic steatosis were used to investigate observations made from human liver tissue. Results: We observed variable degrees of liver steatosis with minimal fibrosis in our participants. Single-cell RNA sequencing revealed four macrophage clusters that exist in the human fatty liver encompassing Kupffer cells and monocyte-derived macrophages (MdMs). The genes expressed in these macrophage subsets were similar to those observed in mouse models of MAFLD. Hepatic CD14+ monocyte/macrophage number correlated with the degree of steatosis. Using mouse models of early liver steatosis, we demonstrate that recruitment of MdMs precedes Kupffer cell loss and liver damage. Electron microscopy of isolated macrophages revealed increased lipid accumulation in MdMs, and ex vivo lipid transfer experiments suggested that MdMs may serve a distinct role in lipid uptake during MAFLD. Conclusions: The human liver in MAFLD contains macrophage subsets that align well with those that appear in mouse models of fatty liver disease. Recruited myeloid cells correlate well with the degree of liver steatosis in humans. MdMs appear to participate in lipid uptake during early stages of MALFD. Impact and implications: Metabolic dysfunction associated fatty liver disease (MAFLD) is extremely common; however, the early inflammatory responses that occur in human disease are not well understood. In this study, we investigated macrophage heterogeneity in human livers during early MAFLD and demonstrated that similar shifts in macrophage subsets occur in human disease that are similar to those seen in preclinical models. These findings are important as they establish a translational link between mouse and human models of disease, which is important for the development and testing of new therapeutic approaches for MAFLD.

6.
Cell Metab ; 34(10): 1499-1513.e8, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36070756

ABSTRACT

Adipocytes transfer mitochondria to macrophages in white and brown adipose tissues to maintain metabolic homeostasis. In obesity, adipocyte-to-macrophage mitochondria transfer is impaired, and instead, adipocytes release mitochondria into the blood to induce a protective antioxidant response in the heart. We found that adipocyte-to-macrophage mitochondria transfer in white adipose tissue is inhibited in murine obesity elicited by a lard-based high-fat diet, but not a hydrogenated-coconut-oil-based high-fat diet, aging, or a corn-starch diet. The long-chain fatty acids enriched in lard suppress mitochondria capture by macrophages, diverting adipocyte-derived mitochondria into the blood for delivery to other organs, such as the heart. The depletion of macrophages rapidly increased the number of adipocyte-derived mitochondria in the blood. These findings suggest that dietary lipids regulate mitochondria uptake by macrophages locally in white adipose tissue to determine whether adipocyte-derived mitochondria are released into systemic circulation to support the metabolic adaptation of distant organs in response to nutrient stress.


Subject(s)
Adipose Tissue, White , Antioxidants , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Antioxidants/metabolism , Diet, High-Fat , Fatty Acids/metabolism , Macrophages/metabolism , Mice , Mitochondria/metabolism , Obesity/metabolism , Starch/metabolism
7.
STAR Protoc ; 2(2): 100511, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33997821

ABSTRACT

Recently, it has become evident that macrophage diversity increases in the liver during the pathogenesis of non-alcoholic steatohepatitis (NASH). Here, we provide a detailed protocol for the analysis of liver macrophage subsets in mice with non-alcoholic fatty liver disease (NAFLD) and early NASH using flow cytometry and immunofluorescence (IF). These methods can be used to assess the composition and localization of macrophage subsets during NASH. For complete details on the use and execution of this protocol, please refer to Daemen et al. (2021).


Subject(s)
Flow Cytometry , Liver , Macrophages , Non-alcoholic Fatty Liver Disease , Animals , Disease Models, Animal , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology
8.
Cell Rep ; 34(2): 108626, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33440159

ABSTRACT

Macrophage-mediated inflammation is critical in the pathogenesis of non-alcoholic steatohepatitis (NASH). Here, we describe that, with high-fat, high-sucrose-diet feeding, mature TIM4pos Kupffer cells (KCs) decrease in number, while monocyte-derived Tim4neg macrophages accumulate. In concert, monocyte-derived infiltrating macrophages enter the liver and consist of a transitional subset that expresses Cx3cr1/Ccr2 and a second subset characterized by expression of Trem2, Cd63, Cd9, and Gpmnb; markers ascribed to lipid-associated macrophages (LAMs). The Cx3cr1/Ccr2-expressing macrophages, referred to as C-LAMs, localize to macrophage aggregates and hepatic crown-like structures (hCLSs) in the steatotic liver. In C-motif chemokine receptor 2 (Ccr2)-deficient mice, C-LAMs fail to appear in the liver, and this prevents hCLS formation, reduces LAM numbers, and increases liver fibrosis. Taken together, our data reveal dynamic changes in liver macrophage subsets during the pathogenesis of NASH and link these shifts to pathologic tissue remodeling.


Subject(s)
Macrophages/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Tissue Engineering/methods , Animals , Disease Models, Animal , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL