scholarly journals PROP1-Dependent Retinoic Acid Signaling Regulates Developmental Pituitary Morphogenesis and Hormone Expression

Endocrinology ◽  
2020 ◽  
Vol 161 (2) ◽  
Author(s):  
Leonard Y M Cheung ◽  
Sally A Camper

Abstract Dietary vitamin A is metabolized into bioactive retinoic acid (RA) in vivo and regulates the development of many embryonic tissues. RA signaling is active in the oral ectoderm-derived tissues of the neuroendocrine system, but its role there has not yet been fully explored. We show here that RA signaling is active during pituitary organogenesis and dependent on the pituitary transcription factor Prop1. Prop1-mutant mice show reduced expression of the aldehyde dehydrogenase gene Aldh1a2, which metabolizes the vitamin A–intermediate retinaldehyde into RA. To elucidate the specific function of RA signaling during neuroendocrine development, we studied a conditional deletion of Aldh1a2 and a dominant-negative mouse model of inhibited RA signaling during pituitary organogenesis. These models partially phenocopy Prop1-mutant mice by exhibiting embryonic pituitary dysmorphology and reduced hormone expression, especially thyrotropin. These findings establish the role of RA in embryonic pituitary stem cell progression to differentiated hormone cells and raise the question of gene-by-environment interactions as contributors to pituitary development and disease.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1009-1009
Author(s):  
Haixia Niu ◽  
Gayla Hadwiger ◽  
Hideji Fujiwara ◽  
John S. Welch

Abstract Introduction: Retinoid receptors are nuclear hormone receptors which are dynamically regulated during terminal myeloid maturation. Retinoic acid receptor α (RARA) is the target of at least ten fusion proteins that lead to acute promyelocytic leukemia (APL). All trans-retinoic acid (ATRA) has been thought to be the principle natural ligand for RARs and it has been used for the treatment of patients with APL. However, the enzymatic pathways that regulate natural retinoids metabolism in hematopoietic cells have not been well defined. ATRA is synthesized from vitamin A through two sequential steps. Vitamin A is oxidized by an alcohol dehydrogenase to yield retinal, which is then irreversibly oxidized by an aldehyde dehydrogenase (ALDH) to generate retinoic acid (RA). At least 19 different human ALDHs have been identified. Among them, ALDH1A1, ALDH1A2 and ALDH1A3 have been shown to oxidize all trans-retinal to ATRA with high affinity, which can be inhibited by diethylaminobenzaldehyde (DEAB). Whether other ALDHs participate in RA metabolism is unknown. Our study identified two distinct retinoid metabolism pathways that are active in bone marrow (BM) progenitors and in macrophages (Mφ). Methods: Gal4-UAS reporter system was used to detect natural RARA ligands in mouse primary hematopoietic cells. We transduced UAS-GFP mouse BM cells with retrovirus that expresses a fusion protein containing the DNA binding domain of Gal4 (which recognizes the UAS promoter) and the ligand binding domain of RARA. An IRES mCherry cassette was included for normalization purposes. Transduced cells were cultured in vitro, or transplanted into lethally irradiated recipient mice. Cells with intracellular RARA ligands activate GFP expression. GFP and mCherry expression were evaluated by flow cytometry. Real-time PCR and Affymetrix array were used to quantify ALDH expression. We identified RARA ligands by mass spectrometry (MS). Results: In vitro, we found that both mouse BM Kit+ cells (Kit+ progenitors) and BM-derived macrophages (BMMφ) could synthesize active RARA ligands via different pathways, but only when the cell culture media was supplemented with vitamin A. Kit+ progenitors utilize DEAB-sensitive ALDH pathways, whereas BMMφ use DEAB-insensitive pathways. By real-time PCR we found Kit+ progenitors have high expression of Aldh1a1, Aldh1a2 and Aldh1a3, whereas BMMφ have no detectable expression of these enzymes. We compared gene expression in Kit+ progenitors and BMMφ by Affymetrix profiling and found that Aldh3b1 was overexpressed in BMMφ. Ectopic expression of Aldh3b1 in 293T cells resulted activation of the same GFP reporter, which could be abrogated by two different antagonist, Ro41-5253 and BMS493, suggesting that Aldh3b1 generated an RARA specific ligand, which we subsequently identified as ATRA via MS. Reciprocally, we found that siRNA knock down of Aldh3b1 in BMMφ reduced the transactivation of the RARA-dependent GFP reporter. The X-RARA fusions have been proposed to act via dominant-negative mechanisms, decreasing retinoid-dependent transcription and myeloid maturation. Surprisingly, in vivo, only rare GFPdim cells were observed in BM cells and no GFP positive cells in peritoneal Mφ of UAS-GFP/Gal4-RARA transplant mice. As positive control, we treated mice with ATRA and observed a dose-dependent GFP increase in both cell types, suggesting that the in vivo reporter can respond to ATRA, but ATRA is not synthesized during adult hematopoiesis, and that dominant-negative inhibition of ATRA-dependent transcription may not be the predominant pathogenic effect of the X-RARA fusion oncoproteins. Conclusion: We have found that at least two distinct enzymatic pathways may be utilized in primary hematopoietic cells to synthesize active RARA ligands from vitamin A. Mouse BM Kit+ progenitors predominantly employ a set of DEAB-sensitive enzymes (Aldh1a1, Aldh1a2 and Aldh1a3), whereas Mφ utilize DEAB-insensitive pathways. We identified Aldh3b1 as a likely candidate and shown that it is capable of ATRA synthesis in vitro. Although these enzymes are expressed in primary BM cells, we found that this does not result in active intracellular RARA ligands in monocytes or Mφ in vivo, suggesting that the rate-limiting step in retinoid synthesis in vivo is likely to involve additional enzymes required for intracellular transport of protein-bound, serum-available vitamin A. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. LBA-4-LBA-4
Author(s):  
Nina Cabezas-Wallscheid ◽  
Florian Buettner ◽  
Daniel Klimmeck ◽  
Pia Sommerkamp ◽  
Luisa Ladel ◽  
...  

Abstract Long-term quiescence or dormancy preserves the genomic integrity as well as the long-term self-renewal and functional capacities of hematopoietic stem cells (HSCs) during homeostasis. In response to infections, inflammatory or chemotherapy induced stress, dormant HSCs (dHSCs) become reversibly activated and are critical for the re-establishment of homeostasis. In our previous work, we defined the molecular landscape of HSCs and its immediate progenitors by determining their DNA-methylome, RNA- transcriptome and their proteome (Cabezas-Wallscheid et al., Cell Stem Cell 2014). This revealed the vitamin A/retinoic acid (RA) signaling pathway to be molecularly predominantly enriched in HSCs. However, the functional relevance of dietary vitamin A for maintenance of HSCs remains uncertain. Moreover, the molecular identity of very rare dHSCs as well as the mechanism regulating their maintenance or the transition out and back into dormancy remains unknown. We now show by single-cell RNA-seq analysis of >300 dHSCs and active HSCs (aHSCs) that the molecular transition from the most inactive dHSCs cluster to the most active HSCs can be best described as a continuous stream-like process linked to a steadily increasing metabolic activation. These single cell derived data are not consistent with a binary switch model, but instead suggest that activation/ differentiation downstream of dHSCs occurs in a continuum without the generation of discrete progenitor cell types. During this process,protein synthesis is increased first, followed by the increase of cell cycle related components. We then measured the time to first division starting from either a dHSC or an aHSC for 285 SiCs by single cell live cell imaging. We found that aHSCs showed an average of 29.5±0.7 hours to enter mitosis, while dHSCs needed 40.8±1.3 hours. This pronounced difference (11.3 hours) between two initially non-cycling populations suggests that dHSCs reside in a deeper level of quiescence, namely dormancy, which is also consistent with the molecular data mentioned above. The association of delayed cell cycle entry with the extremely low biosynthetic activity defines the status of dormancy and distinguishes it from quiescence. Furthermore, based on the acquired expression signatures, we describe the first marker-based, non-label retaining mouse model to specify dHSCs (Gpr-EGFP). We show molecularly and functionally that HSC-Gpr-pos cells resemble dHSCs demonstrating that the Gpr-EGFP mouse line can now be used as a simple alternative approach to track dHSCs and thus circumvent time-consuming label-retaining assays. The Gpr-EGFP model now allows to closely follow cell cycle dynamics within the dHSC compartment. Importantly, the mechanism regulating maintenance and the transition out of dormancy remains unknown. Our data focusing specifically on the most primitive HSCs revealed a critical role for vitamin A/RA signaling in controlling the cell cycle plasticity of dHSCs. We now show by in vitro and in vivo experiments, that treatment with the RA agonist all-trans retinoic-acid (ATRA) preserves dHSCs and maintains critical properties of HSCs. This includes maintenance of long-term self-renewal, low proliferation associated with decreased levels of Cdk6, expression of key transcription factors (Hoxb4), reduced protein synthesis and low levels of reactive oxygen species (ROS) as well as low Myc protein levels. Indeed, in response to activation signals, the presence of ATRA prevents up-regulation of c-Myc protein in HSCs and the effects of ATRA or drug induced Myc inhibition result in similar consequences on HSCs. Moreover, ATRA not only represses ROS production, but also prevents HSCs from entering the cell cycle upon diverse stress stimuli (pIC, LPS, 5-FU) in vivo. Most of the studies on vitamin A deficit-associated immunodeficiency are dedicated to the impaired function of lymphocytes. Thus, we analyzed the consequences of a vitamin A deficient diet for dormant HSCs. Strikingly, we found that HSCs are progressively lost over time and dHSCs did not recover after pIC-mediated activation in the absence of vitamin A. Collectively, these data uncover a critical role of vitamin A/RA signaling for the re-establishment of the dormant HSC population after stress-mediated activation. Together, our results highlight a so far unrecognized impact of dietary vitamin A on the regulation of cell cycle mediated stem cell plasticity. Disclosures No relevant conflicts of interest to declare.


2011 ◽  
Vol 25 (6) ◽  
pp. 908-921 ◽  
Author(s):  
Laura Fozzatti ◽  
Changxue Lu ◽  
Dong-Wook Kim ◽  
Sheue-yann Cheng

Abstract Studies using mice deficient in thyroid hormone receptors (TR) indicate that the two TR isoforms, TRα1 and TRβ1, in addition to mediating overlapping biological activities of the thyroid hormone, T3, also mediate distinct functions. Mice harboring an identical dominant negative mutation (denoted PV) at the C terminus of TRα1 (Thra1PV mice) or β1 (ThrbPV mice) also exhibit distinct phenotypes. These knockin mutant mice provide an opportunity to understand the molecular basis of isoform-dependent functions in vivo. Here we tested the hypothesis that the distinct functions of TR mutant isoforms are directed by a subset of nuclear regulatory proteins. Tandem-affinity chromatography of HeLa nuclear extracts showed that distinct 33 nuclear proteins including nuclear receptor corepressor (NCoR1) and six other proteins preferentially associated with TRα1PV or TRβ1PV, respectively. These results indicate that recruitment of nuclear regulatory proteins by TR mutants is subtype dependent. The involvement of NCoR1 in mediating the distinct liver phenotype of Thra1PV and ThrbPV mice was further explored. NCoR1 preferentially interacted with TRα1PV rather than with TRβ1PV. NCoR1 was recruited more avidly to the thyroid hormone response element-bound TRα1PV than to TRβ1PV in the promoter of the CCAAT/enhancer-binding protein α gene to repress its expression in the liver of Thra1PV mice, but not in ThrbPV mice. This preferential recruitment of NCoR1 by mutant isoforms could contribute, at least in part, to the distinct liver lipid phenotype of these mutant mice. The present study highlights a novel mechanism by which TR isoforms direct their selective functions via preferential recruitment of a subset of nuclear coregulatory proteins.


Development ◽  
1991 ◽  
Vol 111 (4) ◽  
pp. 1081-1086 ◽  
Author(s):  
A.B. Glick ◽  
B.K. McCune ◽  
N. Abdulkarem ◽  
K.C. Flanders ◽  
J.A. Lumadue ◽  
...  

We report the results of a histochemical study, using polyclonal antipeptide antibodies to the different TGF beta isoforms, which demonstrates that retinoic acid regulates the expression of TGF beta 2 in the vitamin A-deficient rat. Basal expression of TGF beta 2 diminished under conditions of vitamin A deficiency. Treatment with retinoic acid caused a rapid and transient induction of TGF beta 2 and TGF beta 3 in the epidermis, tracheobronchial and alveolar epithelium, and intestinal mucosa. Induction of TGF beta 1 expression was also observed in the epidermis. In contrast to these epithelia, expression of the three TGF beta isoforms increased in vaginal epithelium during vitamin A deficiency, and decreased following systemic administration of retinoic acid. Our results show for the first time the widespread regulation of TGF beta expression by retinoic acid in vivo, and suggest a possible mechanism by which retinoics regulate the functions of both normal and pre-neoplastic epithelia.


Development ◽  
1980 ◽  
Vol 59 (1) ◽  
pp. 325-339
Author(s):  
T. E. Kwasigroch ◽  
D. M. Kochhar

Two techniques were used to examine the effect of vitamin A compounds (vitamin A acid = retinoic acid and vitamin A acetate) upon the relative strengths of adhesion among mouse limb-bud mesenchymal cells. Treatment with retinoic acid in vivo and with vitamin A acetate in vitro reduced the rate at which the fragments of mesenchyme rounded-up when cultured on a non-adhesive substratum, but these compounds did not alter the behavior of tissues tested in fragment-fusion experiments. These conflicting results indicate that the two tests measure different activities of cells and suggest that treatment with vitamin A alters the property(ies) of cells which regulate the internal viscosity of tissues.


Blood ◽  
1997 ◽  
Vol 89 (12) ◽  
pp. 4282-4289 ◽  
Author(s):  
Wenlin Shao ◽  
Laura Benedetti ◽  
William W. Lamph ◽  
Clara Nervi ◽  
Wilson H. Miller

Abstract The unique t(15; 17) of acute promyelocytic leukemia (APL) fuses the PML gene with the retinoic acid receptor α (RARα) gene. Although retinoic acid (RA) inhibits cell growth and induces differentiation in human APL cells, resistance to RA develops both in vitro and in patients. We have developed RA-resistant subclones of the human APL cell line, NB4, whose nuclear extracts display altered RA binding. In the RA-resistant subclone, R4, we find an absence of ligand binding of PML-RARα associated with a point mutation changing a leucine to proline in the ligand-binding domain of the fusion PML-RARα protein. In contrast to mutations in RARα found in retinoid-resistant HL60 cells, in this NB4 subclone, the coexpressed RARα remains wild-type. In vitro expression of a cloned PML-RARα with the observed mutation in R4 confirms that this amino acid change causes the loss of ligand binding, but the mutant PML-RARα protein retains the ability to heterodimerize with RXRα and thus to bind to retinoid response elements (RAREs). This leads to a dominant negative block of transcription from RAREs that is dose-dependent and not relieved by RA. An unrearranged RARα engineered with this mutation also lost ligand binding and inhibited transcription in a dominant negative manner. We then found that the mutant PML-RARα selectively alters regulation of gene expression in the R4 cell line. R4 cells have lost retinoid-regulation of RXRα and RARβ and the RA-induced loss of PML-RARα protein seen in NB4 cells, but retain retinoid-induction of CD18 and CD38. Thus, the R4 cell line provides data supporting the presence of an RARα-mediated pathway that is independent from gene expression induced or repressed by PML-RARα. The high level of retinoid resistance in vitro and in vivo of cells from some relapsed APL patients suggests similar molecular changes may occur clinically.


2001 ◽  
Vol 391 (1) ◽  
pp. 8-15 ◽  
Author(s):  
Reza Zolfaghari ◽  
Christopher J. Cifelli ◽  
Melanie D. Banta ◽  
A.Catharine Ross

2006 ◽  
Vol 203 (4) ◽  
pp. 821-828 ◽  
Author(s):  
Hiromichi Matsushita ◽  
Pier Paolo Scaglioni ◽  
Mantu Bhaumik ◽  
Eduardo M. Rego ◽  
Lu Fan Cai ◽  
...  

The promyelocytic leukemia–retinoic acid receptor α (PML-RARα) protein of acute promyelocytic leukemia (APL) is oncogenic in vivo. It has been hypothesized that the ability of PML-RARα to inhibit RARα function through PML-dependent aberrant recruitment of histone deacetylases (HDACs) and chromatin remodeling is the key initiating event for leukemogenesis. To elucidate the role of HDAC in this process, we have generated HDAC1–RARα fusion proteins and tested their activity and oncogenicity in vitro and in vivo in transgenic mice (TM). In parallel, we studied the in vivo leukemogenic potential of dominant negative (DN) and truncated RARα mutants, as well as that of PML-RARα mutants that are insensitive to retinoic acid. Surprisingly, although HDAC1-RARα did act as a bona fide DN RARα mutant in cellular in vitro and in cell culture, this fusion protein, as well as other DN RARα mutants, did not cause a block in myeloid differentiation in vivo in TM and were not leukemogenic. Comparative analysis of these TM and of TM/PML−/− and p53−/− compound mutants lends support to a model by which the RARα and PML blockade is necessary, but not sufficient, for leukemogenesis and the PML domain of the fusion protein provides unique functions that are required for leukemia initiation.


Author(s):  
Frida Ponthan ◽  
Per Borgstr�m ◽  
Moustapha Hassan ◽  
Erik Wassberg ◽  
Christopher P.F. Redfern ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document