EHA Library - The official digital education library of European Hematology Association (EHA)

1 INTRAVASCULAR HEMOLYSIS INDUCES COMPLEMENT SYSTEM ACTIVATION MEDIATED BY HEME AND HEME-LOADED ERYTHROCYTE MICROVESICLES
Author(s): ,
Lubka Roumenina
Affiliations:
INSERM UMRS1138 Cordeliers Research Center,INSERM UMRS1138 Cordeliers Research Center,Paris,France
,
Nicolas Merle
Affiliations:
INSERM UMRS1138 Cordeliers Research Center,INSERM UMRS1138 Cordeliers Research Center,Paris,France
,
Veronique Fremeaux-Bacchi
Affiliations:
HEGP,APHP,paris,France
Olivier Blanc-Brude
Affiliations:
Paris Center for Cardiovascular Research, INSERM UMR_S 970, F-75015, Paris, France,Paris Center for Cardiovascular Research, INSERM UMR_S 970, F-75015, Paris, France,Paris,France
(Abstract release date: 05/17/18) EHA Library. Roumenina L. 06/17/18; 214589; S1587
Dr. Lubka Roumenina
Dr. Lubka Roumenina
Contributions
Abstract

Abstract: S1587

Type: Oral Presentation

Presentation during EHA23: On Sunday, June 17, 2018 from 08:30 - 08:45

Location: Room A9

Background
In hemolytic diseases like sickle cell disease (SCD), intravascular hemolysis results in the release of hemoglobin, heme and heme-loaded membrane microvesicles in the bloodstream. Intravascular hemolysis is then associated with inflammation and organ injury. Our previous studies have demonstrated that the complement system can be activated by heme in vitro. However, the interrelation of intravascular hemolysis, complement activation and tissue damage remains unknown. 

Aims
Our objective was to investigate the mechanisms by which hemolysis and red blood cells degradation products trigger complement activation in vivo

Methods

We evaluated the deposits of C3 activation fragments and C5b-9 in kidneys of SCD patients (n=11) and two transgenic mouse models (SAD and Townes) by immunofluorescence and immunohistochemistry. Intravascular hemolysis was induced in C57Bl/6 mice by injection of phenylhydrazine in presence or not of hemopexin. The complement deposits in the kidneys were followed. RBC microvesicles were generated in vitro from erythrocytes of SCD patients or healthy donors. Complement activation (Ba, sC5b-9) in normal human serum was measured by ELISA. Complement deposits on cultured endothelial cells and their activation status was evaluated by flow cytometry.

Results

We analyzed kidney biopsies of patients with SCD nephropathy and model mice with SCD. We detected significant tissue deposits of complement C3 and C5b-9. Moreover, drug-induced intravascular hemolysis or injection of heme in mice triggered C3 deposition, primarily in the kidneys. Red blood cells degradation products like heme-loaded microvesicles and free heme induced alternative and terminal complement pathway activation in normal serum and on EC surfaces, contrary to hemoglobin. Heme triggered rapid P-selectin, C3aR and C5aR expression and down-regulated CD46 expression on EC. Importantly, complement deposition was attenuated in vivo and in vitro by hemopexin, the potent heme scavenger. 

 

Conclusion
In conclusion, we describe for the first time that intravascular hemolysis triggers complement activation in vivo, and is associated with SCD nephropathyConversely, heme inhibition using hemopexin may provide a novel therapeutic opportunity to limit complement activation in hemolytic diseases.

Session topic: 27. Sickle cell disease

Keyword(s): Complement, heme, Microvesicles, sickle cell disease

Abstract: S1587

Type: Oral Presentation

Presentation during EHA23: On Sunday, June 17, 2018 from 08:30 - 08:45

Location: Room A9

Background
In hemolytic diseases like sickle cell disease (SCD), intravascular hemolysis results in the release of hemoglobin, heme and heme-loaded membrane microvesicles in the bloodstream. Intravascular hemolysis is then associated with inflammation and organ injury. Our previous studies have demonstrated that the complement system can be activated by heme in vitro. However, the interrelation of intravascular hemolysis, complement activation and tissue damage remains unknown. 

Aims
Our objective was to investigate the mechanisms by which hemolysis and red blood cells degradation products trigger complement activation in vivo

Methods

We evaluated the deposits of C3 activation fragments and C5b-9 in kidneys of SCD patients (n=11) and two transgenic mouse models (SAD and Townes) by immunofluorescence and immunohistochemistry. Intravascular hemolysis was induced in C57Bl/6 mice by injection of phenylhydrazine in presence or not of hemopexin. The complement deposits in the kidneys were followed. RBC microvesicles were generated in vitro from erythrocytes of SCD patients or healthy donors. Complement activation (Ba, sC5b-9) in normal human serum was measured by ELISA. Complement deposits on cultured endothelial cells and their activation status was evaluated by flow cytometry.

Results

We analyzed kidney biopsies of patients with SCD nephropathy and model mice with SCD. We detected significant tissue deposits of complement C3 and C5b-9. Moreover, drug-induced intravascular hemolysis or injection of heme in mice triggered C3 deposition, primarily in the kidneys. Red blood cells degradation products like heme-loaded microvesicles and free heme induced alternative and terminal complement pathway activation in normal serum and on EC surfaces, contrary to hemoglobin. Heme triggered rapid P-selectin, C3aR and C5aR expression and down-regulated CD46 expression on EC. Importantly, complement deposition was attenuated in vivo and in vitro by hemopexin, the potent heme scavenger. 

 

Conclusion
In conclusion, we describe for the first time that intravascular hemolysis triggers complement activation in vivo, and is associated with SCD nephropathyConversely, heme inhibition using hemopexin may provide a novel therapeutic opportunity to limit complement activation in hemolytic diseases.

Session topic: 27. Sickle cell disease

Keyword(s): Complement, heme, Microvesicles, sickle cell disease

By clicking “Accept Terms & all Cookies” or by continuing to browse, you agree to the storing of third-party cookies on your device to enhance your user experience and agree to the user terms and conditions of this learning management system (LMS).

Cookie Settings
Accept Terms & all Cookies