Abstract 636: The Nuclear Receptor FXR Uncouples the Actions of Srebp2 and MiR-33

2014 ◽  
Vol 34 (suppl_1) ◽  
Author(s):  
Elizabeth Vallim ◽  
Hannah Ahn ◽  
Thomas Vallim

The role of microRNAs in fine-tuning gene expression has now been well established, but the mechanisms for microRNA regulation at the transcriptional level remain understudied. The physiologic context in which microRNAs are acting is likely linked to the function of these same microRNAs and how they are regulated. Here we identify miR-33 as a direct target gene of the bile acid nuclear receptor FXR. We show that pharmacologic FXR activation induces miR-33 in wild-type, but not FXR knockout mice. MiR-33 is located in the Srebp2 locus, and we show that FXR activation also induces the expression of Srebp2 mRNA. Using ChIP-Seq, we identify at least one functional FXRE in an intronic region of Srebp2, demonstrating that miR-33 and Srebp2 are both direct FXR targets. Although FXR activation increases Srebp2 mRNA levels, we also demonstrate that FXR in fact decreases Srebp2 proteolytic processing, which determines its transcriptional activity. This decreased activity is a result of the FXR dependent induction of the integral ER membrane protein Insig2, which is itself also a direct FXR target. Together these results identify a molecular mechanism for the transcriptional regulation of Srebp2 and miR-33, as well as the mechanism for the decreased activity of Srebp2 processing in the ER by FXR. The best characterized target of miR-33 is ABCA1. We have previously demonstrated that FXR activation decreases ABCA1 protein levels via the induction of miR-144, a microRNA that also targets ABCA1. Our new findings suggest that FXR induces two independent microRNAs that function together to repress ABCA1 and reduce HDL levels.

2013 ◽  
Vol 451 (3) ◽  
pp. 453-461 ◽  
Author(s):  
Claudia C. S. Chini ◽  
Carlos Escande ◽  
Veronica Nin ◽  
Eduardo N. Chini

The nuclear receptor Rev-erbα has been implicated as a major regulator of the circadian clock and integrates circadian rhythm and metabolism. Rev-erbα controls circadian oscillations of several clock genes and Rev-erbα protein degradation is important for maintenance of the circadian oscillations and also for adipocyte differentiation. Elucidating the mechanisms that regulate Rev-erbα stability is essential for our understanding of these processes. In the present paper, we report that the protein DBC1 (Deleted in Breast Cancer 1) is a novel regulator of Rev-erbα. Rev-erbα and DBC1 interact in cells and in vivo, and DBC1 modulates the Rev-erbα repressor function. Depletion of DBC1 by siRNA (small interfering RNA) in cells or in DBC1-KO (knockout) mice produced a marked decrease in Rev-erbα protein levels, but not in mRNA levels. In contrast, DBC1 overexpression significantly enhanced Rev-erbα protein stability by preventing its ubiquitination and degradation. The regulation of Rev-erbα protein levels and function by DBC1 depends on both the N-terminal and C-terminal domains of DBC1. More importantly, in cells depleted of DBC1, there was a dramatic decrease in circadian oscillations of both Rev-erbα and BMAL1. In summary, our data identify DBC1 as an important regulator of the circadian receptor Rev-erbα and proposes that Rev-erbα could be involved in mediating some of the physiological effects of DBC1.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 211-211
Author(s):  
Geqiang Li ◽  
Kristy L. Miskimen ◽  
Zhengqi Wang ◽  
Xiu Yan Xie ◽  
Jennifer Brenzovich ◽  
...  

Abstract Abstract 211 Phosphorylated signal transducer and activator of transcription 5 (STAT5) can inform diagnosis and clinical outcome in myeloid leukemia. STAT5 may be an important therapeutic target for hematologic disease characterized by constitutively active tyrosine kinases. The most direct evidence that STAT5 is crucial for oncogenic transformation has come from mouse models, which showed that STAT5-deficient hematopoietic cells are resistant to transformation by oncogenic tyrosine kinases, such as TEL-JAK2, TEL-PDGF, or BCR-ABL. The oncogenic role of STAT5 has been recognized and likely extends to other activated kinases such as JAK2, MPL, and FLT3. Although up-regulation of genes such as bcl-2, bcl-XL, mcl-1, D-type cyclins, and myc by activated oncogenic tyrosine kinases has been demonstrated, the specific mechanisms how STAT5 can critically control pre-leukemic expansion in the myeloid lineage have not been defined. We have shown previously that lethal myeloproliferative disease (MPD) in mice mediated by persistently activated STAT5 (STAT5aS711F) requires the N-domain but the mechanism was not defined. We now demonstrate by retrovirally complementing STAT5abnull/null primary mast cells that STAT5a lacking the N-domain (STAT5aΔN) ineffectively protected against cytokine withdrawal-induced cell death relative to wild-type STAT5a. To study the mechanisms for this survival defect, bcl-2 and bcl-XL protein levels were analyzed by Western blot and shown to be greatly reduced. Whether bcl-2 is a direct target gene of STAT5 in native chromatin as validated by chromatin immunoprecipitation (ChIP) has not been previously shown. We identified 7 conserved STAT5 binding sites in the bcl-2 gene and tested these by ChIP. Only one site located within intron 2 was bound by STAT5a and STAT5aΔN in mast cells cultured in IL-3 alone, where bcl-2 mRNA levels were low. STAT5a add-back induced bcl-2 mRNA (10-fold) compared with STAT5aΔN (2- to 3-fold). Interestingly, when STAT5 was absent the mRNA levels of bcl-2 were not reduced when mast cells were grown in the presence of both IL-3 and SCF, despite the virtual absence of bcl-2 protein. MicroRNAs (miRs) are small, noncoding, single-stranded RNAs of ∼22 nucleotides that negatively regulate gene expression at the post-transcriptional level primarily through targeting the 3'-UTR of target mRNAs. There are no current reports of STAT5 mediated miR expression. We found that wild-type STAT5a and STAT5aS711F suppressed accumulation of miR15b/16 in primary mast cells and transduced BaF3 cells. Importantly, we show binding of STAT5 to a conserved STAT5 binding site in the promoter of miR15b/16 by ChIP. We propose that reciprocal induction of bcl-2 mRNA and suppression of miR15b/16 maintained bcl-2 protein levels. We also observed binding of STAT5aΔNS711F at both sites by ChIP, indicating that co-activator and co-repressor interactions with the N-domain determine regulation of bcl-2. To test whether these observations of bcl-2 regulation were physiologically relevant in the setting of activated STAT5 in myeloproliferative disease, retroviral complementation of STAT5abnull/null fetal liver cells was performed and followed by transplantation into lethally-irradiated recipients. Persistently active STAT5aS711F lacking the N-domain (STAT5aΔNS711F) was insufficient to protect c-Kit+Lin−Sca-1+ (KLS) cells from apoptosis as determined by Annexin V/DAPI staining. These cells were unable to induce bcl-2 expression determined by intracellular flow cytometry 15 days following injection of donor cells. In contrast, STAT5aS711F caused robust KLS cell expansion, induction of bcl-2, and 47-fold expansion of peripheral Gr-1+Mac-1+ cells. In the absence of the STAT5 N-domain a mild splenomegaly was observed with 50% of mice surviving greater than 90 days, instead of a rapidly lethal monocytic disease observed when full-length STAT5aS711F was present, with death of all mice by 35 days. Importantly, the modest 6-fold increase in peripheral Gr-1+Mac-1+ counts and better survival conferred by STAT5aΔNS711F could be reversed to 41-fold above control levels by adding back only bcl-2 through H2k/bcl-2 transgenic expression. Overall, these studies define N-domain dependent survival signaling as an Achilles' heel of persistent STAT5 activation and highlight the potential therapeutic importance of targeting STAT5 N-domain mediated regulation of bcl-2 family members. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 323-323
Author(s):  
Paola Neri ◽  
Kathy Gratton ◽  
Li Ren ◽  
Jordan Johnson ◽  
Jiri Slaby ◽  
...  

Abstract Abstract 323 Background: miRNAs are non-coding small RNAs that modulate protein expression at the post-transcriptional level and are implicated in the pathogenesis of a variety of cancers. In Multiple Myeloma (MM) a global elevation of miRNAs was previously correlated with poor disease outcomes and response to therapy. Using miRNome profiling of MM patients, we have recently established a miRNA-based risk score that is predictive of response to lenalidomide (Neri P, Blood 2011). In particular, we identified significant upregulation of miR-30 family members (a, b, c and e) in lenalidomide resistant patients. In the present study, we evaluated the biological functions of miR-30e in MM and its role in plasma cells resistance to lenalidomide as well as other anti-MM therapeutics. Methods and Results: Microarray profiling (Affymetrix miRNA GeneChip) of total RNA extracted from bone marrow plasma cells from lenalidomide sensitive and resistant MM patients (n=40), coupled with quantitative short stem-loop PCR (TaqMan, Applied Biosystems), confirmed the upregulation of miR-30e in lenalidomide resistant patients. Functionally, we sought to determine if overexpression of miR-30e would modify MM cells sensitivity to lenalidomide and bortezomib. Lentiviral-mediated stable expression (pLKO.1 retroviral plasmid) of miR-30e, and relative to empty vector (EV), significant increased MM1S and OPM2 cells growth (1.3 fold) as determined by MTT assay. In addition, miR-30e overexpressing cells (MM1S-30e and OPM2-30e vs MM1-EV and OPM2-EV) were more resistant to the cytotoxic effects of lenalidomide as well as bortezomib with approximately 15 to 20% reduction in cells death (Annexin V staining and MTT assay). Computational target prediction analysis (TargetScan 6.0 and miRanda) identified CRBN and BLIMP1 as potential target of miR-30e with a miRNA seed region that matches 8 or 7mer sites within Cereblon and BLIMP1 3'UTR regions. In a panel of MM cell lines (MM1S, OPM2, H929, INA-6, U266, 8226, KMS11) CRBN mRNA levels were indeed inversely correlated with miR-30e and stable mir-30e overexpression significantly reduced CRBN mRNA in these cells (MM1S-30e and OPM2-30e). In addition to CRBN, BLIMP1 mRNA and protein levels were also reduced in miR-30e overexpressing cells. In plasma cells, BLIMP1 drives XBP1 expression while supressing c-myc. In MM1S-30e and OPM2-30e (relative to empty vector), and consistent with their reduced BLIMP1 expression, XBP1 mRNA and protein levels were reduced. Furthermore, treatment with lenalidomide (10μM) significantly reduced c-MYC protein levels in MM1S-EV cells after 4 hours while it had no effect on C-MYC expression in MM1S-30e cells. Conclusions: miR-30e is overexpressed in resistant MM cells and is here shown to regulate cereblon expression, plasma cells differentiation axis (BLIMP1, XBP1) and cell growth (c-MYC). Disclosures: Neri: Johnson ans Johnson: Research Funding. Bahlis:Johnson and Johnson: Honoraria, Research Funding; Celgene: Honoraria.


2020 ◽  
Vol 202 (10) ◽  
Author(s):  
Dharam Singh ◽  
Oleg N. Murashko ◽  
Sue Lin-Chao

ABSTRACT Escherichia coli ribosomal protein (r-protein) L4 has extraribosomal biological functions. Previously, we described L4 as inhibiting RNase E activity through protein-protein interactions. Here, we report that from stabilized transcripts regulated by L4-RNase E, mRNA levels of tnaA (encoding tryptophanase from the tnaCAB operon) increased upon ectopic L4 expression, whereas TnaA protein levels decreased. However, at nonpermissive temperatures (to inactivate RNase E), tnaA mRNA and protein levels both increased in an rne temperature-sensitive [rne(Ts)] mutant strain. Thus, L4 protein fine-tunes TnaA protein levels independently of its inhibition of RNase E. We demonstrate that ectopically expressed L4 binds with transcribed spacer RNA between tnaC and tnaA and downregulates TnaA translation. We found that deletion of the 5′ or 3′ half of the spacer compared to the wild type resulted in a similar reduction in TnaA translation in the presence of L4. In vitro binding of L4 to the tnaC-tnaA transcribed spacer RNA results in changes to its secondary structure. We reveal that during early stationary-phase bacterial growth, steady-state levels of tnaA mRNA increased but TnaA protein levels decreased. We further confirm that endogenous L4 binds to tnaC-tnaA transcribed spacer RNA in cells at early stationary phase. Our results reveal the novel function of L4 in fine-tuning TnaA protein levels during cell growth and demonstrate that r-protein L4 acts as a translation regulator outside the ribosome and its own operon. IMPORTANCE Some ribosomal proteins have extraribosomal functions in addition to ribosome translation function. The extraribosomal functions of several r-proteins control operon expression by binding to own-operon transcripts. Previously, we discovered a posttranscriptional, RNase E-dependent regulatory role for r-protein L4 in the stabilization of stress-responsive transcripts. Here, we found an additional extraribosomal function for L4 in regulating the tna operon by L4-intergenic spacer mRNA interactions. L4 binds to the transcribed spacer RNA between tnaC and tnaA and alters the structural conformation of the spacer RNA, thereby reducing the translation of TnaA. Our study establishes a previously unknown L4-mediated mechanism for regulating gene expression, suggesting that bacterial cells have multiple strategies for controlling levels of tryptophanase in response to varied cell growth conditions.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4451-4451 ◽  
Author(s):  
Nicola Amodio ◽  
Marzia Leotta ◽  
Lavinia Biamonte ◽  
Teresa Calimeri ◽  
Eugenio Morelli ◽  
...  

MicroRNAs (miRNAs) are short non-coding RNAs that control gene expression at the post-transcriptional level by inducing mRNA decay or translation repression. A subclass of miRNAs, named epi-miRNAs, is known to exert anti-tumor activity by targeting effectors of the epigenetic machinery. We recently demonstrated a key role for the tumor suppressor miR-29b in reducing the global DNA methylation of multiple myeloma (MM) cells through the targeting of DNA-methyltransferases. In silico search of additional miR-29b targets contributing to clarify its role as epi-miRNA unveiled the class II histone deacetylase HDAC4. Since histone deacetylases represent promising molecular targets for cancer treatment, we sought to characterize HDAC4 expression and function and its regulation by miR-29b in MM cells. HDAC4 protein levels and enzymatic activity were found up-regulated in a panel of 11 MM cell lines as compared to peripheral blood mononuclear cells from healthy donors. Moreover, the analysis of HDAC4 mRNA levels in a microarray dataset consisting of 4 healthy controls, 55 MM and 29 plasma cell leukemia patients indicated significant over-expression in cancer samples, suggesting that high HDAC4 expression might be involved in MM pathogenesis. Notably, the analysis of miRNA/mRNA paired expression in the same microarray dataset revealed an inverse correlation between miR-29b and HDAC4 mRNA, strengthening the relevance of miR-29b as a key regulator of HDAC4. Synthetic miR-29b mimics transfected in MM cells (SKMM1 and NCI-H929) down-regulated HDAC4 mRNA and protein levels and inhibited the 3’UTR of HDAC4 cloned in a luciferase reporter vector, whereas failed to regulate a 3’UTR devoid of two predicted miR-29b target sites. On the other hand, lentiviral-mediated overexpression of HDAC4 strongly inhibited miR-29b expression. These results underscore a negative feedback loop occurring between miR-29b and its target HDAC4 in MM cells. Through loss of function experiments, we assessed the functional significance of HDAC4 in MM cells. Stable silencing of HDAC4 by shRNAs induced growth inhibition, caspase 3/7-dependent apoptosis and autophagy in U266 and KMS11 cells, which occurred together to miR-29b up-regulation. Interestingly, the pan-HDAC inhibitor vorinostat also triggered apoptosis and autophagy in MM cells, along with the induction of miR-29b and the down-regulation of HDAC4 and other miR-29b-canonical targets like CDK6, MCL-1 and Sp1. miR-29b itself was able to promote autophagy, as assessed by beclin-1 up-regulation and LC3A/B proteolytic cleavage in miR-29b mimics-transfected MM cells, which was abrogated by constitutive expression of HDAC4. Finally, we provided evidence that miR-29b over-expression potentiated, whereas its stable inhibition dampened, apoptosis and autophagy triggered by vorinostat. In conclusion, our findings shed light on the oncogenic role of HDAC4, which can be targeted through miR-29b-based therapeutic approaches, and identify miR-29b as a relevant effector of vorinostat activity in MM cells. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 3 (1) ◽  
pp. e201900513 ◽  
Author(s):  
Shun-saku Takahashi ◽  
Yu-Shin Sou ◽  
Tetsuya Saito ◽  
Akiko Kuma ◽  
Takayuki Yabe ◽  
...  

Lipid droplets (LDs) are dynamic organelles that store neutral lipids during times of energy excess, such as after a meal. LDs serve as an energy reservoir during fasting and have a buffering capacity that prevents lipotoxicity. Autophagy and the autophagic machinery have been proposed to play a role in LD biogenesis, but the underlying molecular mechanism remains unclear. Here, we show that when nuclear receptor co-repressor 1 (NCoR1), which inhibits the transactivation of nuclear receptors, accumulates because of autophagy suppression, LDs decrease in size and number. Ablation of ATG7, a gene essential for autophagy, suppressed the expression of gene targets of liver X receptor α, a nuclear receptor responsible for fatty acid and triglyceride synthesis in an NCoR1-dependent manner. LD accumulation in response to fasting and after hepatectomy was hampered by the suppression of autophagy. These results suggest that autophagy controls physiological hepatosteatosis by fine-tuning NCoR1 protein levels.


2017 ◽  
Vol 2017 ◽  
pp. 1-10 ◽  
Author(s):  
He Peiyuan ◽  
Hou Zhiping ◽  
Song Chengjun ◽  
Wang Chunqing ◽  
Li Bingqing ◽  
...  

The aim of this study was to investigate the hepatoprotective effects of resveratrol in alcoholic liver disease (ALD). Alcohol was administered to healthy female rats starting from 6% (v/v) and gradually increased to 20% (v/v) by the fifth week. After 16 weeks of intervention, liver enzymes (aspartate aminotransferase [AST] and alanine aminotransferase [ALT]) were analyzed using a chemistry analyzer, while hepatic antioxidant enzymes, oxidative stress markers, and caspase 3 activity were assessed using ELISA kits. Furthermore, hepatic CYP2E1 protein levels and mRNA levels of antioxidant and inflammation-related genes were determined using western blotting and RT-PCR, respectively. The results showed that resveratrol significantly attenuated alcohol-induced elevation of liver enzymes and improved hepatic antioxidant enzymes. Resveratrol also attenuated alcohol-induced CYP2E1 increase, oxidative stress, and apoptosis (caspase 3 activity). Moreover, genes associated with oxidative stress and inflammation were regulated by resveratrol supplementation. Taken together, the results suggested that resveratrol alleviated ALD through regulation of oxidative stress, apoptosis, and inflammation, which was mediated at the transcriptional level. The data suggests that resveratrol is a promising natural therapeutic agent against chronic ALD.


2008 ◽  
Vol 389 (3) ◽  
pp. 305-312 ◽  
Author(s):  
Pascal Lovis ◽  
Sonia Gattesco ◽  
Romano Regazzi

Abstract Fine-tuning of insulin secretion from pancreatic β-cells participates in blood glucose homeostasis. Defects in this process can lead to chronic hyperglycemia and diabetes mellitus. Several proteins controlling insulin exocytosis have been identified, but the mechanisms regulating their expression remain poorly understood. Here, we show that two non-coding microRNAs, miR124a and miR96, modulate the expression of proteins involved in insulin exocytosis and affect secretion of the β-cell line MIN6B1. miR124a increases the levels of SNAP25, Rab3A and synapsin-1A and decreases those of Rab27A and Noc2. Inhibition of Rab27A expression is mediated by direct binding to the 3′-untranslated region of Rab27A mRNA. The effect on the other genes is indirect and linked to changes in mRNA levels. Over-expression of miR124a leads to exaggerated hormone release under basal conditions and a reduction in glucose-induced secretion. miR96 increases mRNA and protein levels of granuphilin, a negative modulator of insulin exocytosis, and decreases the expression of Noc2, resulting in lower capacity of MIN6B1 cells to respond to secretagogues. Our data identify miR124a and miR96 as novel regulators of the expression of proteins playing a critical role in insulin exocytosis and in the release of other hormones and neurotransmitters.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1189-1189
Author(s):  
Sakiko Suzuki ◽  
Nathan A Manalo ◽  
Glen D Raffel

Abstract Ott1(Rbm15) is essential for engraftment and maintaining hematopoietic stem cell (HSC) quiescence during proliferative stress; therefore we sought to establish whether Ott1 has a regulatory role within the cell cycle. Ott1 knockout (KO) E14.5 murine embryonic fibroblasts (MEFs) were analyzed using BrdU labelling and demonstrated a higher basal proliferative rate. However, when subjected to oncogenic stress induced by infection with a constitutively active N-Ras expressing retrovirus, Ott1-deleted MEFs undergo immortalization and morphologic transformation in contrast to wild type (WT) MEFs which undergo senescence. Oncogene-induced senescence is a p53-facilitated process. P53 protein levels were shown by western blot to decrease in Ras-infected Ott1 KO MEFs rather than increase as observed in WT Ras-infected MEFs. Consistent with this finding, p16Ink4a, which is a transcriptional target of p53, is not upregulated in Ras-infected Ott1 KO MEFs. Gamma irradiation was still able to induce p53 in Ott1 KO MEFS, demonstrating Ott1 regulation of p53 is specific to the oncogenic stress pathway, but not the DNA damage pathway. Measurement of p53 mRNA levels in Ras-infected Ott1 KO MEFs showed a modest increase compared to WT, indicating the p53 protein decrease must occur at a post-transcriptional level. Classical p53 induction by oncogenic stress occurs through inhibition of ubiquitin-mediated degradation of p53 by ligases such as Mdm2 and Mdm4. To determine why Ras induction of p53 is defective in Ott1 KO MEFS, Ras-infected cells were incubated with the proteasome inhibitor, MG132, which was able to rescue p53 induction, implicating a ubiquitination-dependent mechanism. Furthermore, incubation with Nutlin3, an Mdm2-specific inhibitor, also showed significant rescue of p53 induction, signifying Ott1 is required for Mdm2-mediated degradation of p53 during oncogenic stress. P53 has an essential, non-apoptotic role in HSC function and has also been shown to help maintain HSC quiescence and self-renewal. We previously identified an Ott1-dependent mechanism for down-regulating Thrombopoietin response via its receptor Mpl in Ott1 KO HSCs through expression of a dominant negative alternatively spliced isoform, Mpl-TR. Although Mpl-TR expression is sufficient to reduce Mpl signaling and competitive repopulation in Ott1 KO HSCs, full length Mpl alone is unable to rescue engraftment of Ott1 -deleted HSCs suggesting Ott1 has other critical targets. Based on the Ott1-dependence of p53 function in MEFs, we hypothesized a similar dysfunction of the p53 pathway exists in Ott1 KO HSCs undergoing proliferative stress. Ott1 KO and WT HSCs were analyzed before and after incubation in a cytokine-rich medium to stimulate proliferation. At baseline, Ott1 KO HSCs have similar p53 protein levels as WT HSCs. However, after cytokine stimulation, Ott1 KO HSCs shift into active cell cycle more readily and now demonstrate a significant decrease in p53 protein levels as measured by intracellular flow cytometry. In summary, Ott1 is required for p53 response during oncogenic stress via inhibition of Mdm2. Ott1 is similarly required to maintain p53 levels during proliferative stress in HSCs and may thereby promote quiescence and self-renewal. Moreover, OTT1 is the 5' fusion partner in the chimeric OTT1-MAL (RBM15-MKL1) product in t(1;22)-associated acute megakaryocytic leukemia, raising the possibility that dysregulation of p53 pathways may contribute to the pathogenesis of t(1;22)-derived leukemias. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Author(s):  
Shun-saku Takahashi ◽  
Yu-Shin Sou ◽  
Tetsuya Saito ◽  
Akiko Kuma ◽  
Takayuki Yabe ◽  
...  

AbstractLipid droplets (LDs) are dynamic organelles that store neutral lipids during times of energy excess, such as following a meal. LDs serve as an energy reservoir during fasting and have a buffering capacity that prevents lipotoxicity. Autophagy and the autophagic machinery have been proposed to play a role in LD biogenesis but the underlying molecular mechanism remains unclear. Here, we show that when nuclear receptor co-repressor 1 (NCoR1), which inhibits the transactivation of nuclear receptors, accumulates due to autophagy suppression, LD biogenesis is blocked. Ablation of ATG7, a gene essential for autophagy, suppressed the expression of gene targets of liver X receptor α (LXRα), a nuclear receptor responsible for fatty acid and triglyceride synthesis in an NCoR1-dependent manner. LD biogenesis in response to fasting and after hepatectomy was hampered by the suppression of autophagy. These results indicate that autophagy controls physiological hepatosteatosis by fine-tuning NCoR1 protein levels.


Sign in / Sign up

Export Citation Format

Share Document