The pathogenic role of c-Kit+ mast cells in the spinal motor neuron-vascular niche in ALS


Topic:

Pre-Clinical Research

Poster Number: 116

Author(s):

Emiliano Trías, PhD, Institut Pasteur, Mariángeles Kovacs, Institut Pasteur de Montevideo, Catalina Alamón, Cecilia Maciel, Valentina Varela, Sofía Ibarburu, Institut Pasteur de Montevideo, Lucas Tarragó, Peter H King, UAB Neurology, Ying Si, UAB Neurology, Yuri Kwon, Olivier Hermine, Institut IMAGINE, Hopital Necker, Luis Barbeito, Institut Pasteur de Montevideo

Background
Degeneration of motor neurons, glial cell reactivity, and vascular alterations in the CNS are important neuropathological features of amyotrophic lateral sclerosis (ALS). Immune cells trafficking from the blood also infiltrate the CNS parenchyma and contribute to neuroinflammation. Mast cells (MCs) are hematopoietic-derived immune cells whose precursors differentiate upon migration into tissues. Upon activation, MCs undergo degranulation with the ability to increase vascular permeability, orchestrate neuroinflammation and modulate the neuroimmune response.

Objective
However, the prevalence, pathological significance, and pharmacology of MCs in the CNS of ALS patients remain largely unknown. Here we aim to identify and characterize whether mast cells infiltrate into the neurodegenerative microenvironment that surrounds degenerating motor neurons during the paralysis progression in ALS.

Results
In autopsy ALS spinal cords, we identified for the first time that MCs express c-Kit together with chymase, tryptase, and Cox-2 and display granular or degranulating morphology, as compared with scarce MCs in control cords. In ALS, MCs were mainly found in the niche between spinal motor neuron somas and nearby microvascular elements, and they displayed remarkable pathological abnormalities. Similarly, MCs accumulated in the motor neuron-vascular niche of ALS murine models, in the vicinity of astrocytes and motor neurons expressing the c-Kit ligand stem cell factor (SCF), suggesting an SCF/c-Kit-dependent mechanism of MC differentiation from precursors. Mechanistically, we provide evidence that fully differentiated MCs in cell cultures can be generated from the murine ALS spinal cord tissue, further supporting the presence of c-Kit+ MC precursors. Moreover, intravenous administration of bone marrow-derived c-Kit+ MC precursors infiltrated the spinal cord in ALS mice but not in controls, consistent with aberrant trafficking through a defective microvasculature. Pharmacological inhibition of c-Kit with masitinib in ALS mice reduced the MC number and the influx of MC precursors from the periphery.

Conclusions
Our results suggest a previously unknown pathogenic mechanism triggered by MCs in the ALS motor neuron-vascular niche that might be targeted pharmacologically.