Pulmonary transplantation of human induced pluripotent stem cell–derived macrophages ameliorates pulmonary alveolar proteinosis

C Happle, N Lachmann, M Ackermann… - American Journal of …, 2018 - atsjournals.org
C Happle, N Lachmann, M Ackermann, A Mirenska, G Göhring, K Thomay, A Mucci…
American Journal of Respiratory and Critical Care Medicine, 2018atsjournals.org
Rationale: Although the transplantation of induced pluripotent stem cell (iPSC)–derived cells
harbors enormous potential for the treatment of pulmonary diseases, in vivo data
demonstrating clear therapeutic benefits of human iPSC–derived cells in lung disease
models are missing. Objectives: We have tested the therapeutic potential of iPSC-derived
macrophages in a humanized disease model of hereditary pulmonary alveolar proteinosis
(PAP). Hereditary PAP is caused by a genetic defect of the GM-CSF (granulocyte …
Rationale: Although the transplantation of induced pluripotent stem cell (iPSC)–derived cells harbors enormous potential for the treatment of pulmonary diseases, in vivo data demonstrating clear therapeutic benefits of human iPSC–derived cells in lung disease models are missing.
Objectives: We have tested the therapeutic potential of iPSC-derived macrophages in a humanized disease model of hereditary pulmonary alveolar proteinosis (PAP). Hereditary PAP is caused by a genetic defect of the GM-CSF (granulocyte–macrophage colony–stimulating factor) receptor, which leads to disturbed macrophage differentiation and protein/surfactant degradation in the lungs, subsequently resulting in severe respiratory insufficiency.
Methods: Macrophages derived from human iPSCs underwent intrapulmonary transplantation into humanized PAP mice, and engraftment, in vivo differentiation, and therapeutic efficacy of the transplanted cells were analyzed.
Measurements and Main Results: On intratracheal application, iPSC-derived macrophages engrafted in the lungs of humanized PAP mice. After 2 months, transplanted cells displayed the typical morphology, surface markers, functionality, and transcription profile of primary human alveolar macrophages. Alveolar proteinosis was significantly reduced as demonstrated by diminished protein content and surfactant protein D levels, decreased turbidity of the BAL fluid, and reduced surfactant deposition in the lungs of transplanted mice.
Conclusions: We here demonstrate for the first time that pulmonary transplantation of human iPSC–derived macrophages leads to pulmonary engraftment, their in situ differentiation to an alveolar macrophage phenotype, and a reduction of alveolar proteinosis in a humanized PAP model. To our knowledge, this finding presents the first proof-of-concept for the therapeutic potential of human iPSC-derived cells in a pulmonary disease and may have profound implications beyond the rare disease of PAP.
ATS Journals