Anti-CX3CL1 (fractalkine) monoclonal antibody attenuates lung and skin fibrosis in sclerodermatous graft-versus-host disease mouse model

Arthritis Res Ther. 2024 May 3;26(1):94. doi: 10.1186/s13075-024-03307-8.

Abstract

Background: Systemic sclerosis (SSc) is an autoimmune disease characterized by vascular injury and inflammation, followed by excessive fibrosis of the skin and other internal organs, including the lungs. CX3CL1 (fractalkine), a chemokine expressed on endothelial cells, supports the migration of macrophages and T cells that express its specific receptor CX3CR1 into targeted tissues. We previously reported that anti-CX3CL1 monoclonal antibody (mAb) treatment significantly inhibited transforming growth factor (TGF)-β1-induced expression of type I collagen and fibronectin 1 in human dermal fibroblasts. Additionally, anti-mouse CX3CL1 mAb efficiently suppressed skin inflammation and fibrosis in bleomycin- and growth factor-induced SSc mouse models. However, further studies using different mouse models of the complex immunopathology of SSc are required before the initiation of a clinical trial of CX3CL1 inhibitors for human SSc.

Methods: To assess the preclinical utility and functional mechanism of anti-CX3CL1 mAb therapy in skin and lung fibrosis, a sclerodermatous chronic graft-versus-host disease (Scl-cGVHD) mouse model was analyzed with immunohistochemical staining for characteristic infiltrating cells and RNA sequencing assays.

Results: On day 42 after bone marrow transplantation, Scl-cGVHD mice showed increased serum CX3CL1 level. Intraperitoneal administration of anti-CX3CL1 mAb inhibited the development of fibrosis in the skin and lungs of Scl-cGVHD model, and did not result in any apparent adverse events. The therapeutic effects were correlated with the number of tissue-infiltrating inflammatory cells and α-smooth muscle actin (α-SMA)-positive myofibroblasts. RNA sequencing analysis of the fibrotic skin demonstrated that cGVHD-dependent induction of gene sets associated with macrophage-related inflammation and fibrosis was significantly downregulated by mAb treatment. In the process of fibrosis, mAb treatment reduced cGVHD-induced infiltration of macrophages and T cells in the skin and lungs, especially those expressing CX3CR1.

Conclusions: Together with our previous findings in other SSc mouse models, the current results indicated that anti-CX3CL1 mAb therapy could be a rational therapeutic approach for fibrotic disorders, such as human SSc and Scl-cGVHD.

Keywords: 3 ~ 10; CX3CL1; CXCR1; Fibrosis; Fractalkine; Graft-versus-host disease; Lung; Scleroderma; Skin; Systemic sclerosis.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antibodies, Monoclonal* / pharmacology
  • Antibodies, Monoclonal* / therapeutic use
  • Chemokine CX3CL1* / antagonists & inhibitors
  • Chemokine CX3CL1* / metabolism
  • Disease Models, Animal*
  • Female
  • Fibrosis
  • Graft vs Host Disease* / drug therapy
  • Graft vs Host Disease* / immunology
  • Graft vs Host Disease* / pathology
  • Humans
  • Lung / drug effects
  • Lung / immunology
  • Lung / metabolism
  • Lung / pathology
  • Mice
  • Mice, Inbred C57BL
  • Pulmonary Fibrosis* / drug therapy
  • Pulmonary Fibrosis* / immunology
  • Pulmonary Fibrosis* / pathology
  • Pulmonary Fibrosis* / prevention & control
  • Scleroderma, Systemic* / drug therapy
  • Scleroderma, Systemic* / immunology
  • Scleroderma, Systemic* / pathology
  • Skin* / drug effects
  • Skin* / immunology
  • Skin* / metabolism
  • Skin* / pathology

Substances

  • Chemokine CX3CL1
  • Antibodies, Monoclonal
  • Cx3cl1 protein, mouse