Neonatal bone marrow transplantation (nBMT) could offer a novel therapeutic opportunity for genetic disorders by providing sustainable levels of the missing protein at birth, thus preventing tissue damage. We tested this concept in Mucopolysaccharidosis type I (MPS IH, Hurler syndrome), a lysosomal storage disorder caused by deficiency of α-L-iduronidase (IDUA). MPS IH is characterized by a broad spectrum of clinical manifestations including severe progressive skeletal abnormalities. Although BMT increases the life span of MPS IH patients, musculoskeletal manifestations are only minimally responsive if the timing of BMT delays, suggesting already irreversible bone damage. In this study, we tested the hypothesis that transplanting normal bone marrow into newborn MPS I mice, soon after birth, can prevent skeletal dysplasia. 1- to 2-day-old mutant mice were conditioned using a single administration of 20 mg/kg busulfan and then injected via the superficial temporal vein with 2 x 106 bone marrow cells from wild type (WT) donors. Age-matched WT and untreated MPS I mice were used as controls.

Transplantation of normal bone marrow cells into preconditioned MPS I and WT neonates led to a similar engraftment level at 37 weeks after nBMT (peripheral blood, median MPS I nBMT 81.30%, range from 0.80% to 95.80% vs. median WT nBMT 67.45%, range from 16.00% to 95.86%, p = 0.714). Spleen, PB and thymus cells of nBMT MPS I mice were repopulated with committed lymphoid and myeloid populations similar to the transplanted WT mice. The >50% replacement of the hematopoiesis resulted in a measurable increase in IDUA activity in visceral organs, especially in the spleen, showing a correlation between engraftment levels and enzyme activity with clearance of GAGs from blood and tissues. At the time of euthanasia (37-week-old), reconstitution of normal hematopoiesis in MPS I mice was associated with a consistent amelioration of bone pathology, as revealed by radiographic skeletal examination. Radiographic analysis has shown that the width of the humerus, radius/ulna, femur and tibia of untreated MPS I mice was significantly larger at comparison with WT littermates. For MPS I nBMT mice, long bone widths, including the humerus (p = 0.0014, vs. untreated MPS I mice), the radius/ulna (p = 0.0003, vs. untreated MPS I mice), the femur (p = 0.0003, vs. untreated MPS I mice), and the tibia (p = 0.0003, vs. untreated MPS I mice) significantly decreased, compared to untreated MPS I mice. Furthermore, several three-dimensional architectural parameters in femurs such as trabecular number and separation, cortical thickness and bone mineral volume were analyzed by micro-CT, resulting in a significant difference between untreated and nBMT MPS I mice. All examined nBMT MPS I mice displayed bone parameter values comparable to WT mice, confirming that nBMT mice had significant improvements in skeletal phenotype approaching complete normalization of each parameter tested. Histologically, in MPS I cortical bone, osteocytes were increased and contained vacuoles, likely reflecting GAGs storage. Histological amelioration of these features was consistently observed in femurs of all nBMT mice with a definite reduction in both hyperosteocytosis and lysosomal vacuolization, confirming that the perinatal treatment of the disease can positively impact the skeletal phenotype in MPS I. We also evaluated KS levels in the blood as a biomarker of MPS with skeletal dysplasia. Normalization of blood KS level strongly supports the notion that nBMT corrects the pathological and clinical bone lesions in nBMT MPS I mice. Our findings demonstrate that nBMT prevents some of the relevant abnormalities of the skeletal pathology in the MPS I mouse model. Moreover, improvements in bone parameters correlated with high levels of bone marrow-derived cell engraftment in multiple hematopoietic compartments, suggesting that the early and complete restoration of normal hematopoiesis can have significant impact on the bone development of newborn MPS I mice. Future clinical trials are needed to confirm our findings.

Disclosures

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution