Your browser doesn't support javascript.
loading
Complementary HLH susceptibility factors converge on CD8 T-cell hyperactivation.
Landy, Emily; Varghese, Jemy; Dang, Vinh; Szymczak-Workman, Andrea; Kane, Lawrence P; Canna, Scott W.
Afiliación
  • Landy E; Department of Immunology, University of Pittsburgh, Pittsburgh, PA.
  • Varghese J; Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA.
  • Dang V; Rheumatology & Immune Dysregulation, Children's Hospital of Philadelphia, Philadelphia, PA.
  • Szymczak-Workman A; Rheumatology & Immune Dysregulation, Children's Hospital of Philadelphia, Philadelphia, PA.
  • Kane LP; Department of Immunology, University of Pittsburgh, Pittsburgh, PA.
  • Canna SW; Department of Immunology, University of Pittsburgh, Pittsburgh, PA.
Blood Adv ; 7(22): 6949-6963, 2023 11 28.
Article en En | MEDLINE | ID: mdl-37738167
ABSTRACT
Hemophagocytic lymphohistiocytosis (HLH) and macrophage activation syndrome (MAS) are life-threatening hyperinflammatory syndromes. Familial HLH is caused by genetic impairment of granule-mediated cytotoxicity (eg, perforin deficiency). MAS is linked to excess activity of the inflammasome-activated cytokine interleukin-18 (IL-18). Though individually tolerated, mice with dual susceptibility (Prf1⁻/⁻Il18tg; DS) succumb to spontaneous, lethal hyperinflammation. We hypothesized that understanding how these susceptibility factors synergize would uncover key pathomechanisms in the activation, function, and persistence of hyperactivated CD8 T cells. In IL-18 transgenic (Il18tg) mice, IL-18 effects on CD8 T cells drove MAS after a viral (lymphocytic choriomeningitis virus), but not innate (toll like receptor 9), trigger. In vitro, CD8 T cells also required T-cell receptor (TCR) stimulation to fully respond to IL-18. IL-18 induced but perforin deficiency impaired immunoregulatory restimulation-induced cell death (RICD). Paralleling hyperinflammation, DS mice displayed massive postthymic oligoclonal CD8 T-cell hyperactivation in their spleens, livers, and bone marrow as early as 3 weeks. These cells increased proliferation and interferon gamma production, which contrasted with increased expression of receptors and transcription factors associated with exhaustion. Broad-spectrum antibiotics and antiretrovirals failed to ameliorate the disease. Attempting to genetically "fix" TCR antigen-specificity instead demonstrated the persistence of spontaneous HLH and hyperactivation, chiefly on T cells that had evaded TCR fixation. Thus, drivers of HLH may preferentially act on CD8 T cells IL-18 amplifies activation and demand for RICD, whereas perforin supplies critical immunoregulation. Together, these factors promote a terminal CD8 T-cell activation state, combining features of exhaustion and effector function. Therefore, susceptibility to hyperinflammation may converge on a unique, unrelenting, and antigen-dependent state of CD8 T-cell hyperactivation.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Linfohistiocitosis Hemofagocítica Límite: Animals Idioma: En Revista: Blood Adv Año: 2023 Tipo del documento: Article País de afiliación: Panamá

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Linfohistiocitosis Hemofagocítica Límite: Animals Idioma: En Revista: Blood Adv Año: 2023 Tipo del documento: Article País de afiliación: Panamá