Your browser doesn't support javascript.
loading
Single-cell profiling of intrahepatic immune cells reveals an expansion of tissue-resident cytotoxic CD4+ T lymphocyte subset associated with pathogenesis of alcoholic-associated liver diseases.
Gao, Chao; Wang, Shiguan; Xie, Xiaoyu; Ramadori, Pierluigi; Li, Xinying; Liu, Xiaoyu; Ding, Xue; Liang, Jinyuan; Xu, Bowen; Feng, Yawei; Tan, Xueying; Wang, Haoran; Zhang, Yan; Zhang, Haiyan; Zhang, Tingguo; Mi, Ping; Li, Shiyang; Zhang, Cuijuan; Yuan, Detian; Heikenwalder, Mathias; Zhang, Peng.
Afiliação
  • Gao C; Department of Hepatobiliary Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Wang S; Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Xie X; Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China.
  • Ramadori P; Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome
  • Li X; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Liu X; Advanced Medical Research Institute, Shandong University, Jinan, Shandong 250012, China.
  • Ding X; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Liang J; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Xu B; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Feng Y; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Tan X; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Wang H; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Zhang Y; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Zhang H; Department of Biochemistry, Heze Medical College, Heze, Shandong 274000, China.
  • Zhang T; Institute of Pathology and Pathophysiology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Mi P; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
  • Li S; Advanced Medical Research Institute, Shandong University, Jinan, Shandong 250012, China.
  • Zhang C; Institute of Pathology and Pathophysiology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pathology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China. Electronic address: cuijuanzhang@sdu.edu.cn.
  • Yuan D; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China. Electronic address: yuandt@sdu.edu.cn.
  • Heikenwalder M; Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome
  • Zhang P; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China. Electronic address: zhangpeng1990@sdu.edu.cn.
Cell Mol Gastroenterol Hepatol ; : 101411, 2024 Sep 28.
Article em En | MEDLINE | ID: mdl-39349248
ABSTRACT
BACKGROUND &

AIMS:

The immunological mechanisms underpinning the pathogenesis of alcoholic-associated liver disease (ALD) remain incompletely elucidated. This study aims to explore the transcriptomic profiles of hepatic immune cells in ALD compared to healthy individuals and those with metabolic dysfunction-associated steatotic liver disease (MASLD).

METHODS:

We utilized single-cell RNA sequencing to analyze liver samples from healthy subjects, MASLD, and ALD patients, focusing on the immune cell landscapes within the liver. Key alterations in immune cell subsets were further validated using liver biopsy samples from additional patient cohorts.

RESULTS:

We observed a significant accumulation of CD4+ T cells in ALD patients' livers, surpassing the prevalence of CD8+ T cells, in contrast to MASLD and healthy counterparts, while natural killer (NK) cells and γδT cells exhibited reduced intrahepatic infiltration. In-depth transcriptional and developmental trajectory analyses unveiled that a distinct CD4+ subset characterized by granzyme K (GZMK) expression, displaying a tissue-resident signature and terminal effector state, prominently enriched among CD4+ T cells infiltrating the livers of ALD patient. Subsequent examination of an independent ALD patient cohort corroborated the substantial enrichment of GZMK+CD4+ T lymphocytes, primarily within liver fibrotic zones, suggesting their potential involvement in disease progression. Additionally, we noted shifts in myeloid populations, with expanded APOE+ macrophage and FCGR3B+ monocyte subsets in ALD samples relative to MASLD and healthy tissues.

CONCLUSIONS:

In summary, this study unravels the intricate cellular diversity within hepatic immune cell populations, highlighting the pivotal immune pathogenic role of the GZMK+CD4+ T lymphocyte subset in ALD pathogenesis.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article