Your browser doesn't support javascript.
loading
Steatosis drives monocyte-derived macrophage accumulation in human metabolic dysfunction-associated fatty liver disease.
Chan, Mandy M; Daemen, Sabine; Beals, Joseph W; Terekhova, Marina; Yang, Bin Q; Fu, Christina F; He, Li; Park, Arick C; Smith, Gordon I; Razani, Babak; Byrnes, Kathleen; Beatty, Wandy L; Eckhouse, Shaina R; Eagon, J Christopher; Ferguson, Daniel; Finck, Brian N; Klein, Samuel; Artyomov, Maxim N; Schilling, Joel D.
Affiliation
  • Chan MM; Diabetes Research Center, Washington University School of Medicine, St. Louis, MO, USA.
  • Daemen S; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
  • Beals JW; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
  • Terekhova M; Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands.
  • Yang BQ; Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA.
  • Fu CF; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
  • He L; Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
  • Park AC; Diabetes Research Center, Washington University School of Medicine, St. Louis, MO, USA.
  • Smith GI; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
  • Razani B; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
  • Byrnes K; Diabetes Research Center, Washington University School of Medicine, St. Louis, MO, USA.
  • Beatty WL; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
  • Eckhouse SR; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
  • Eagon JC; Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA.
  • Ferguson D; Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA.
  • Finck BN; Division of Cardiology, Pittsburgh VA Medical Center, Pittsburgh, PA, USA.
  • Klein S; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
  • Artyomov MN; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
  • Schilling JD; Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
JHEP Rep ; 5(11): 100877, 2023 Nov.
Article in En | 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.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: JHEP Rep Year: 2023 Document type: Article Affiliation country: United States Publication country: HOLANDA / HOLLAND / NETHERLANDS / NL / PAISES BAJOS / THE NETHERLANDS

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: JHEP Rep Year: 2023 Document type: Article Affiliation country: United States Publication country: HOLANDA / HOLLAND / NETHERLANDS / NL / PAISES BAJOS / THE NETHERLANDS