4.7 Article

RhoG deficiency abrogates cytotoxicity of human lymphocytes and causes hemophagocytic lymphohistiocytosis

Journal

BLOOD
Volume 137, Issue 15, Pages 2033-2045

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/blood.2020008738

Keywords

-

Categories

Funding

  1. European Research Council through ERC [820074]
  2. Vienna Science and Technology Fund [LS14-031]
  3. Austrian Academy of Science through DOC Fellowships [25365, 25225]
  4. Finnish Foundation for Pediatric Research and Pediatric Research Center, Helsinki University Hospital
  5. Swedish Research Council
  6. Cancer Foundation
  7. Children's Cancer Foundation
  8. Knut and Alice Wallenberg Foundation
  9. European Research Council (ERC) [820074] Funding Source: European Research Council (ERC)

Ask authors/readers for more resources

This study identifies RhoG as a novel essential regulator in aiding the retention of cytotoxic granules near the plasma membrane, facilitating their release for the elimination of infected and malignant cells. The interaction between RhoG and Munc13-4 is crucial for the docking of cytotoxic granules to the plasma membrane and subsequent membrane fusion, shedding light on the molecular pathomechanism behind genetically determined forms of hemophagocytic lymphohistiocytosis.
Exocytosis of cytotoxic granules (CG) by lymphocytes is required for the elimination of infected and malignant cells. Impairments in this process underly a group of diseases with dramatic hyperferritinemic inflammation termed hemophagocytic lymphohistiocytosis (HLH). Although genetic and functional studies of HLH have identified proteins controlling distinct steps of CG exocytosis, the molecular mechanisms that spatiotemporally coordinate CG release remain partially elusive. We studied a patient exhibiting characteristic clinical features of HLH associated with markedly impaired cytotoxic T lymphocyte (CTL) and natural killer (NK) cell exocytosis functions, who beared biallelic deleterious mutations in the gene encoding the small GTPase RhoG. Experimental ablation of RHOG in a model cell line and primary CTLs from healthy individuals uncovered a hitherto unappreciated role of RhoG in retaining CGs in the vicinity of the plasma membrane (PM), a fundamental prerequisite for CG exocytotic release. We discovered that RhoG engages in a protein-protein interaction with Munc13-4, an exocytosis protein essential for CG fusion with the PM. We show that this interaction is critical for docking of Munc13-41 CGs to the PM and subsequent membrane fusion and release of CG content. Thus, our study illuminates RhoG as a novel essential regulator of human lymphocyte cytotoxicity and provides the molecular pathomechanism behind the identified here and previously unreported genetically determined form of HLH.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available