, 2012) Reprogramming technologies, such as iPSC or iN generatio

, 2012). Reprogramming technologies, such as iPSC or iN generation, theoretically “erase” the existing epigenetic state of a cell and establish an alternative state. Such epigenetic states are determined in part by direct modifications of genomic DNA, including methylation or hydroxymethylation, as well as by binding of chromatin factors such as histones that modify selleck screening library the accessibility of genomic DNA (Tomazou and Meissner, 2010). Yet other regulators,

that include both protein and non-coding RNA factors, serve to refine the epigenetic state of individual genetic loci. Additionally, the three-dimensional structure of chromatin, determined by yet poorly defined nuclear elements, may selleck chemical broadly impact the epigenetic program. In the context of patient-derived cultures, historical events of potential relevance to disease—such

as aging or toxin exposure—may theoretically underlie a persistent change in epigenetic state, and this may in turn impact cellular phenotypes. The cell-type-specific epigenetic state of a starting cell—in contrast to genetic factors—is predicted to be “erased” in the context of somatic cell reprogramming. Thus, epigenetic reprogramming models, such as patient iPSC-derived neurons, may not display a given disease phenotype, if it is epigenetic in origin. Conversely, a disease-associated phenotype that is apparent in reprogramming-derived cell models is predicted to be genetic in origin. A caveat is that reprogramming has often appeared incomplete: “epigenetic memory” persists in iPSC-derived cultures as to their cells of origin (Kim et al., 2010 and Kim et al., 2011c) as well as with directed reprogramming

(Khachatryan et al., 2011). Going forward, it will be of high interest to directly assess epigenetic Bumetanide changes associated with disease states in reprogrammed neuron models. In some contexts, “incomplete reprogramming—which retains significant epigenetic memory—may be desirable. More speculatively, directed reprogramming to neurons may present an advantage over iPSC reprogramming followed by differentiation; single step reprogramming to neurons is perhaps more likely to retain epigenetic memory of prior events, leading to disease-related cellular phenotypes. However, epigenetic memory in skin cells may not be relevant to CNS disorders. In summary, the application of reprogramming technologies toward the generation of accurate and simple human cell models of adult neurological disorders is a promising approach. It is perhaps unexpected that diseases of aging such as familial Alzheimer’s disease would be recapitulated to some extent “in a dish.” This reflects an emerging theme, in which underlying molecular and cellular culprits to these diseases of aging may often be present throughout life, whereas unknown “second hits” ultimately lead to the full expression of disease.

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>