Antibiotics impair murine hematopoiesis by depleting the intestinal microbiota

KS Josefsdottir, MT Baldridge… - Blood, The Journal of …, 2017 - ashpublications.org
KS Josefsdottir, MT Baldridge, CS Kadmon, KY King
Blood, The Journal of the American Society of Hematology, 2017ashpublications.org
Bone marrow suppression is an adverse effect associated with many antibiotics, especially
when administered for prolonged treatment courses. Recent advances in our understanding
of steady-state hematopoiesis have allowed us to explore the effects of antibiotics on
hematopoietic progenitors in detail using a murine model. Antibiotic-treated mice exhibited
anemia, thrombocytosis, and leukopenia, with pronounced pan-lymphopenia as
demonstrated by flow cytometric analysis of peripheral blood. Bone marrow progenitor …
Abstract
Bone marrow suppression is an adverse effect associated with many antibiotics, especially when administered for prolonged treatment courses. Recent advances in our understanding of steady-state hematopoiesis have allowed us to explore the effects of antibiotics on hematopoietic progenitors in detail using a murine model. Antibiotic-treated mice exhibited anemia, thrombocytosis, and leukopenia, with pronounced pan-lymphopenia as demonstrated by flow cytometric analysis of peripheral blood. Bone marrow progenitor analysis revealed depletion of hematopoietic stem cells and multipotent progenitors across all subtypes. Granulocytes and B cells were also diminished in the bone marrow, whereas the number of CD8+ T cells increased. Reductions in progenitor activity were not observed when cells were directly incubated with antibiotics, suggesting that these effects are indirect. Hematopoietic changes were associated with a significant contraction of the fecal microbiome and were partially rescued by fecal microbiota transfer. Further, mice raised in germ-free conditions had hematopoietic abnormalities similar to those seen in antibiotic-treated mice, and antibiotic therapy of germ-free mice caused no additional abnormalities. The effects of antibiotics were phenocopied in Stat1-deficient mice, with no additional suppression by antibiotics in these mice. We conclude that microbiome depletion as a result of broad-spectrum antibiotic treatment disrupts basal Stat1 signaling and alters T-cell homeostasis, leading to impaired progenitor maintenance and granulocyte maturation. Methods to preserve the microbiome may reduce the incidence of antibiotic-associated bone marrow suppression.
ashpublications.org