Abstract 1003

Introduction:

The Congenital dyserythropoietic anemias (CDAs) are a heterogeneous group of rare inborn disorders mainly affecting erythropoiesis. Distinct from other inherited bone marrow failure syndromes, they are marked by morphological abnormalities of the erythroblasts leading to ineffective erythropoiesis/dyserythropoiesis, while the other hematopoietic lineages appear to be unaffected. Based largely on the dysplastic changes observed in erythroblasts by light and electron microscopy, the CDAs have been divided into 3 major types (CDA-1, 2 and 3). Causative genes have been identified for CDA-1 (CDAN1/15q15) and CDA-2 (CDAN2/20p11), while the chromosomal locus is known for CDA-3 (15q22). The majority of CDA-1 cases are associated with CDAN1 missense mutations, leading to single amino-acid substitutions in the 134 kDa Codanin-1 protein. Electron microscopy of CDA-1 erythroblasts reveals a characteristic pattern of spongy (“Swiss cheese”) heterochromatin. We investigated the chromatin in CDA-1 patients, the localization, distribution and interactions of Codanin-1 and generated a murine knock-out model for CDAN1.

Results:

Given the grossly abnormal chromatin ultrastructure in CDA-1 erythroblasts, we examined its overall content. No gross differences between normal and patient samples were seen, both in the amounts of histone proteins or various epigenetic marks of histone tails, suggesting that histone signatures, involved in maintenance of chromatin structure and epigenetic regulation, are globally maintained CDA-1. We confirmed the latter by ChIP-on-chip analysis where immunoprecipitated H4 acetylated, H3K4 dimethylated, H3K9 trimethylated and H3K27 trimethylated chromatin from CDA-1 erythroblasts and normal controls was hybridized to ENCODE genome-wide microrrays. Using a hybridoma technique, we produced 3 novel monoclonal antibodies to Codanin-1. These consistently demonstrate its distribution in both nucleus and cytoplasm of cells by immunofluorescence and Western Blotting. While Codanin-1 is found in the nucleus, it is more abundant in the cytoplasm in primary human erythroblasts. This localization pattern was unchanged in CDA-1 erythroblasts. Within the nucleus, Codanin-1 is localized in foci, while in the cytoplasm it shows an aggregated distribution, with the endoplasmic reticulum and Golgi apparatus being the most likely locations. No differences were found in the distribution patterns of RNA-polymerase-II, PML, nucleophosmin, HP1β and HP1γ between patients' and control erythroblasts. However, the localization of HP1α, a key component of heterochromatin, was found to be markedly perturbed. HP1α accumulates in the Golgi apparatus of CDA-1 but not normal erythroblasts. This was not seen in lymphoblasts derived from CDA-1 patients, nor in erythroblasts from a patient with CDA-2. We confirmed that the abnormal localization of HP1α in CDA-1 patients is confined to the intermediate erythroblast maturation stage, where the characteristic ultrastructural chromatin pattern of CDA-1 is observed. Immunoprecipitation of erythroblast protein extracts with anti- HP1α antibodies co-precipitated Codanin-1, suggesting that an abnormality in Codanin-1 could be responsible for the aberrant localization of HP1α. By confocal immunofluorescence, we also found Codanin-1 to co-localize partially with SEC23B, the protein mutated in CDA-2, suggesting a molecular link between the two major types of CDA. We generated the first murine Cdan1 gene-trap model demonstrating its widespread expression during embryonic development. Cdan1gt/gt homozygotes die in-utero before the onset of primitive erythropoiesis, suggesting that Cdan1 has other critical roles during embryogenesis. Adult heterozygous Cdan1wt/gt mice had normal hematopoietic function and morphology, normal life expectancy and fertility. No extra-hematopoietic macroscopic phenotype could be identified.

Conclusions:

Our results show that the absence of the highly conserved and ubiquitously expressed Codanin-1 is embryonic lethal, and suggest that missense CDAN1 mutations are responsible for the erythroid specific phenotype of CDA-1 via the abnormal cellular trafficking of the heterochromatin protein HP1α. Moreover, our findings suggest that common molecular pathways underlie the CDAs.

Disclosures:

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution