Interestingly, when testing mRNAs from the FACS-purified hrGFP cells on a specially designed qPCR array for 93 different peptide precursors, we observed that the transcript for the neuropeptide adenylate cyclase-activating polypeptide 1 (encoding PACAP) was highly enriched (150-fold) followed by secretin (80-fold)

Interestingly, when testing mRNAs from the FACS-purified hrGFP cells on a specially designed qPCR array for 93 different peptide precursors, we observed that the transcript for the neuropeptide adenylate cyclase-activating polypeptide 1 (encoding PACAP) was highly enriched (150-fold) followed by secretin (80-fold). -cells, and a fraction of pancreatic polypeptide cells. It is concluded that ChgA-hrGFP in the GI tract functions as an effective reporter, particularly for the large populations of still poorly characterized monoamine-storing enteroendocrine cells. Furthermore, our findings substantiate the potential function of ChgA as a monoamine-binding protein that facilitates the regulated endocrine secretion of large amounts of monoamines from enteroendocrine cells. Chromogranin A (ChgA) is an acidic glycoprotein found in large dense-core vesicles of Pyridoxine HCl the regulated secretory pathway (1, 2). A number of other vesicular proteins sharing physiochemical properties with ChgA have been identified, giving rise to a family of granins, which includes chromogranin B (ChgB), secretogranin II, secretogranin III, HISL-19 antigen (SgIV), 7B2 (SgV), NESP55 (SgVI), VGF nerve factor inducible (VGF, SgVII), and pro-SAAS (SgVIII) (3). Although many different functions have been proposed for granins and granin-derived peptides, their mechanisms of action and physiological importance in most cases still remain to be elucidated (3). ChgA and other granins are thought to act as precursors for smaller peptide fragments; however, potential receptors, downstream signaling pathways, and biological activities remain unknown (3, 4). They have also been implicated in the actual formation of the large, dense-core secretory vesicles and in the sorting of proteins to the regulated secretory pathway (5). Most convincingly, the large, acidic ChgA protein has been proposed to function as an osmotic regulator, binding large amounts of solutes to facilitate the accumulation of monoamines within the dense-core secretory vesicles of the endocrine cells. This function as a storage capacity generating protein has in particular been advocated for ChgA in the adrenaline-storing chromaffin cells of the adrenal medulla (2, 6). ChgA specifically localizes to peptide hormone and monoamine-storing endocrine cells within the gastrointestinal (GI) tract and was therefore early on accepted as a general marker for enteroendocrine cells as such (7,C10), which is still often used as. However, although colocalization of ChgA and enteroendocrine peptide hormones has been shown to a variable degree for nearly all known gut hormones, a large degree of heterogeneity has been noted among species and GI Pyridoxine HCl regions (11, 12). Technical variations between studies aside, differential posttranslational processing of the ChgA proprotein in different cell types could contribute to the observed differences in ChgA colocalization (13, 14). In contrast to the inconsistent overlap between ChgA and peptide hormone expression, ChgA has consistently been associated with enteroendocrine cells producing and secreting monoamines, ie, the histamine producing enterochromaffin (EC)-like (ECL) cells of the stomach and the serotonin-producing EC cells, which constitute a major population of enteroendocrine cells throughout the GI tract (11, 12). Notably, gut-derived serotonin has recently been demonstrated to function as a true endocrine factor that modulates adipose and hepatic function (15). Furthermore, the so-called peripheral serotonin, which is almost entirely derived from the EC cells, has been shown also to control the functions of brown adipose tissue (16,C18). Several transgenic reporter mice have been generated for individual enteroendocrine cell types based on the transcriptional control elements for peptide hormone precursors, transcription factors and chemosensors (19, 20). However, a reporter mouse based on a general marker protein would enable characterization of the total enteroendocrine cell population. Thus, in the present study, we have generated a transgenic reporter mouse expressing humanized (hr) green fluorescent protein (GFP), under the control Antxr2 of the transcriptional control elements for ChgA. The reporter protein was found to be expressed in ChgA storing cells of the adrenal medulla, the pituitary, the endocrine pancreas and throughout the GI tract. Interestingly, in the GI tract, hrGFP was expressed mainly in monoamine-storing cells, which makes the reporter mouse a particularly useful resource for future studies of these important, but still poorly characterized, cells. Materials and Methods Production of ChgA-hrGFP transgenic mice A ChgA-containing bacterial artificial chromosome (BAC) (bMQ421c07) was chosen based on sequence information from Pyridoxine HCl www.ensembl.org and obtained from the BACPAC Resources Center at Children’s Hospital Oakland Research Institute. The ChgA BAC contained 87 447 base pairs upstream from the.