Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • 2024-04
  • 2024-05
  • br Introduction S http www apexbt com

    2024-05-06


    Introduction Systemic lupus erythematosus (SLE) is a group of systemic autoimmune disorders characterized by anti-nuclear Melanocyte stimulating hormone release inhibiting factor (ANA), rashes and photosensitivity, joint inflammation, nephritis, and other clinical criteria. SLE develops through the breakdown of three major checkpoints: adaptive immune tolerance, peripheral innate immune responsiveness, and end-organ inflammation [1]. Adaptive immune dysfunction produces autoantibodies leading to immune complex formation and deposition in the skin, joints, and kidneys. Innate immunity plays an important role in determining disease severity and progression [2]. Factors and pathways that modulate innate immunity impact the course of disease, particularly in end organ systems where most major, life-threatening disease manifestations occur. We focus here on the Axl receptor tyrosine kinase pathway. Axl is a member of the TAM family of receptor tyrosine kinases. Axl engagement by its ligand Gas6 causes receptor homodimerization, autophosphorylation, and downstream signaling. Axl and other TAM family members are well-studied in malignancy but also play significant roles in immunity [3], [4], [5], [6], [7], [8]. Among immune cells, Axl mRNA expression is highest in macrophages. Axl expression is also reported in some dendritic cells, γδT cells, fibroblasts, CD25+ T cells, and B1a B cells [9]. Disturbance of TAM family function contributes to autoimmunity. TAM family triple knockout mice develop severe systemic autoimmunity, and two members have been implicated directly in autoimmunity [10]. The TAM family member Mer aids in the clearance of apoptotic cells that may otherwise exacerbate inflammation and autoimmunity [11], [12]. Further, Axl knockout mice exhibit worse inflammation and demyelination than normal controls in induced experimental autoimmune encephalomyelitis (EAE) [13]. Two major functions of Axl may influence SLE. First, Gas6-stimulated Axl in the kidney is necessary for renal mesangial cell proliferation, which contributes to nephritis [14], [15], [16], [17]. Second, Axl signaling blocks the expression of inflammatory cytokines through the induction of Twist, a transcriptional inhibitor, in macrophages [18]. We and others quantified soluble Axl (sAxl) in the serum of SLE patients and healthy controls [19], [20], [21]. Patients with active SLE exhibit significantly elevated sAxl versus healthy controls and patients with inactive SLE. Others have proposed modulation of the Axl tyrosine kinase pathway as a new target for SLE, but no feasible mechanism has been determined [22]. We hypothesized that leukocyte Axl is cleaved in lupus, abrogating anti-inflammatory signaling and contributing to the disease.
    Materials and methods
    Results
    Discussion and conclusion In the present study we observe that SLE patients and lupus-prone mice exhibit increased levels of sheared soluble Axl ectodomain (sAxl) in the blood and reduced surface Axl and active Y779-phosphorylated Axl on immune cells. This occurs despite increased Axl ligand Gas6 in SLE serum [19], [20], [21]. We demonstrate that matrix metalloproteases ADAM10 and TACE (ADAM17) cleave surface Axl on CD11b+ or CD14+ and CD19+ lupus leukocytes. Under normal conditions, Axl acts to transduce anti-inflammatory signals in macrophages through Twist-mediated suppression of inflammatory cytokines [18]. This represents a part of the innate immune checkpoint, and dysregulation of this system may contribute to the pathology of autoimmune disease. We demonstrate that the loss of surface Axl abrogates Axl-mediated anti-inflammatory activity in vitro and in vivo. Both Axl-deficient and lupus-prone mouse macrophages fail to induce Twist or block expression of IL-6 and TNF-alpha in response to Gas6. Axl-deficient BMDM worsen end organ damage in vivo. To our knowledge these are the first data to show the functional significance of Melanocyte stimulating hormone release inhibiting factor Axl ectodomain shearing in lupus.