Ji-Hye Paik's research while affiliated with Weill Cornell Medical College and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (71)


Mitochondria Define Intestinal Stem Cell Differentiation Downstream of a FOXO/Notch Axis
  • Article

November 2020

·

58 Reads

·

94 Citations

Cell Metabolism

Marlies C Ludikhuize

·

Maaike Meerlo

·

Marc Pages Gallego

·

[...]

·

Differential WNT and Notch signaling regulates differentiation of Lgr5+ crypt-based columnar cells (CBCs) into intestinal cell lineages. Recently we showed that mitochondrial activity supports CBCs, while adjacent Paneth cells (PCs) show reduced mitochondrial activity. This implies that CBC differentiation into PCs involves a metabolic transition toward downregulation of mitochondrial dependency. Here we show that Forkhead box O (FoxO) transcription factors and Notch signaling interact in determining CBC fate. In agreement with the organoid data, Foxo1/3/4 deletion in mouse intestine induces secretory cell differentiation. Importantly, we show that FOXO and Notch signaling converge on regulation of mitochondrial fission, which in turn provokes stem cell differentiation into goblet cells and PCs. Finally, scRNA-seq-based reconstruction of CBC differentiation trajectories supports the role of FOXO, Notch, and mitochondria in secretory differentiation. Together, this points at a new signaling-metabolic axis in CBC differentiation and highlights the importance of mitochondria in determining stem cell fate.

Share

Rapid and Selective Targeting of Heterogeneous Pancreatic Neuroendocrine Tumors
  • Article
  • Full-text available

March 2020

·

98 Reads

·

8 Citations

iScience

Design of tissue-specific contrast agents to delineate tumors from background tissues is a major unmet clinical need for ultimate surgical interventions. Bioconjugation of fluorophore(s) to a ligand has been mainly used to target overexpressed receptors on tumors. However, the size of the final targeted ligand can be large >20 kDa and cannot readily cross the microvasculature to meet the specific tissue, resulting in low targetability with a high background. Here, we report a small and hydrophilic phenoxazine with high targetability and retention to pancreatic neuroendocrine tumor. This bioengineered fluorophore permits sensitive detection of ultrasmall (<0.5 mm) ectopic tumors within a few seconds after a single bolus injection, highlighting every tumor in the pancreas from the surrounding healthy tissues with reasonable half-life. The knowledge-based approach and validation used to develop structure-inherent tumor targeted fluorophores have a tremendous potential to improve treatment outcome by providing definite tumor margins for image-guided surgery.

Download

Mitochondria define intestinal stem cell differentiation downstream of a FOXO/Notch axis

September 2019

·

104 Reads

·

1 Citation

Differential signalling of the WNT and Notch pathways regulates proliferation and differentiation of Lgr5+ crypt-based columnar cells (CBCs) into all cell lineages of the intestine. We have recently shown that high mitochondrial activity in CBCs is key in maintaining stem cell function. Interestingly, while high mitochondrial activity drives CBCs, it is reduced in the adjacent secretory Paneth cells (PCs). This observation implies that during differentiation towards PCs, CBCs undergo a metabolic rewiring involving downregulation of mitochondrial number and activity, through a hitherto unknown mechanism. Here we demonstrate, using intestinal organoids that FoxO transcription factors and Notch signalling functionally interact in determining CBC cell fate. In agreement with the organoid data, combined Foxo1 and 3 deletion in mice increases PC number in the intestine. Importantly, we show that FOXO and Notch signalling converge onto regulation of mitochondrial fission, which in turn provokes stem cell differentiation into the secretory types; Goblet cells and PCs. Finally, mapping intestinal stem cell differentiation based on pseudotime computation of scRNA-seq data further supports the role of FOXO, Notch and mitochondria in determining secretory differentiation. This shows that mitochondria is not only a discriminatory hallmark of CBCs and PCs, but that its status actively determines lineage commitment during differentiation. Together, our work describes a new signalling-metabolic axis in stem cell differentiation and highlights the importance of mitochondria in determining cell fate.


FoxO1 regulates leptin-induced mood behavior by targeting tyrosine hydroxylase

November 2018

·

63 Reads

·

4 Citations

Metabolism

Purpose While leptin has been associated with various psycho-physiological functions, the molecular network in leptin-mediated mood regulation remains elusive. Methods Anxiolytic behaviors and tyrosine hydroxylase (TH) levels were examined after leptin administration. Functional roles of STAT3 and FoxO1 in regulation of TH expression were investigated using in vivo and in vitro systems. A series of animal behavioral tests using dopaminergic neuron-specific FoxO1 KO (FoxO1 KODAT) were performed and investigated the roles of FoxO1 in regulation of mood behaviors. Results Here, we show that administration of leptin induces anxiolytic-like phenotype through the activation of signal transducer and activator of transcription 3 (STAT3) and the inhibition of forkhead box protein O1 (FoxO1) in dopaminergic (DA) neurons of the midbrain. Specifically, STAT3 and FoxO1 directly bind to and exert opposing effects on tyrosine hydroxylase (TH) expression, where STAT3 acts as an enhancer and FoxO1 acts as a prominent repressor. Accordingly, suppression of the prominent suppressor FoxO1 by leptin strongly increased TH expression. Furthermore, our previous results showed that specific deletion of FoxO1 in DA neurons (FoxO1 KODAT) led to a profound elevation of TH activity and dopamine contents. Finally, FoxO1 KODAT mice exhibited enhanced leptin sensitivity as well as displayed reduced anxiety- and depression-like behaviors. Conclusions This work establishes a novel molecular mechanism of mood behavior regulation by leptin and suggests FoxO1 suppression by leptin might be a key for leptin-induced behavioral manifestation in DA neurons.



Figure 1: Generation of DA neuron-specific FoxO1 KO (FoxO1 KODAT) mice.: (a) DA neuron-specific FoxO1 deletion. Green fluorescence indicates FoxO1. TdTomato (Td, red) was used to visualize DA neurons expressing DAT-cre. DAPI stains nuclei (blue). Scale bar, 50 μm. (b) Allele-specific PCR using different brain areas from FoxO1F/+, FoxO1F/F, and FoxO1 KODAT (DAT-cre; FoxO1F/F). SN, substantia nigra. Hypo., hypothalamus. Cb, cerebellum. (c,d) Immunoblots (c) and relative FoxO1 protein levels (d) from indicated brain regions of FoxO1 KODAT and WT littermates. (e) Brain weight of FoxO1 KODAT and WT littermates. (f) Representative figures and graphs showing the brain width of FoxO1 KODAT and WT littermates. Scale bar, 10 mm. (g–i) Representative figures (g) and graphs showing the number (h) and size (i) of DA neurons of FoxO1 KODAT and WT littermates. Scale bar, 50 μm. The values are mean±s.e.m. (*P<0.05, **P<0.01, Student’s t-test).
Figure 2: Metabolic phenotypes of FoxO1 KODAT mice in chow diet.: (a) Body weight of male mice on chow diet. Body weight data were combined from three cohorts of mice. (b) GTT and area under curve (AUC) of 24-week old male mice fed chow diet. (c) Plasma insulin levels of male mice during GTT (Not significant from two-way analysis of variance (ANOVA)). (d) ITT and AUC of 24-week-old male mice on chow diet. The results are expressed as mean±s.e.m. (*P<0.05, Student’s t-test for bar graphs and two-way ANOVA for comparison of multiple time points).
Figure 3: FoxO1 KODAT mice are resistant to diet-induced obesity.: (a) Body weight change (%) after HFD feeding (HFD started at 8 weeks old). Body weight data were combined from 4 cohorts. (b) Body fat and lean mass of male mice at 6 weeks on HFD. (c,d) Serum insulin (c) and leptin (d) levels of male mice at 6 weeks on HFD (Body weight, WT: 38.93±1.75 g, KO: 34.95±1.63 g, P=0.13 for c and WT: 37.87±1.78 g, KO: 33.92±1.50 g, P=0.12 for d, respectively). (e) Blood glucose (fed) level of male mice at 10 weeks on HFD (Body weight, WT: 42.71±2.12 g and KO: 36.18±2.17 g, P=0.04). (f) GTT and AUC of male mice on HFD for 10 weeks (Body weight, WT: 37.94±2.07 g and KO: 32.74±1.31 g, P=0.04). (g) ITT and AUC of male mice on HFD for 10 weeks (Body weight, WT: 39.64±1.96 g and KO: 33.86±1.55 g, P=0.03). The results are expressed as mean±s.e.m. (*P<0.05, Student’s t-test for bar graphs and two-way analysis of variance for comparison of multiple time points in line graphs).
Figure 4: Feeding behaviour of FoxO1 KODAT mice.: (a) Daily food intake of 8-10 weeks old male mice on chow diet (averaged from 7 days). (b–d) Rebound food intake (b) rebound weight gain (c) and rebound blood glucose (d) of overnight fasted male mice after re-feeding with normal chow. (e) Percentage of sucrose preference in fed and fasted conditions of male mice. 2% sucrose was used for the preference test. (f) Daily HFD consumption of 8-10 weeks old male mice (averaged from 7 days). The results are expressed as mean±s.e.m. (*P<0.05, Student’s t-test for bar graphs in a,e–f and two-way analysis of variance for comparison of multiple time points in b–d).
Figure 5: Increased energy expenditure in FoxO1 KODAT mice.: (a) Cumulative food intake of male mice fed on HFD for 1 week. (b) Locomotor activity. (c) Temporal changes of O2 consumption. (d) Average O2 consumption. (e) Temporal changes of CO2 production. (f) Average CO2 production. (g) Heat generation between genotypes. (h) Rectal temperature of male and female mice measured at room temperature environment. ♂, male. ♀, female. NS, not significant. Data are expressed as mean±s.e.m. (*P<0.05, Student’s t-test for bar graphs in h and two-way analysis of variance for multiple comparisons in a–g).

+2

FoxO1 in dopaminergic neurons regulates energy homeostasis and targets tyrosine hydroxylase

September 2016

·

421 Reads

·

38 Citations

Nature Communications

Dopaminergic (DA) neurons are involved in the integration of neuronal and hormonal signals to regulate food consumption and energy balance. Forkhead transcriptional factor O1 (FoxO1) in the hypothalamus plays a crucial role in mediation of leptin and insulin function. However, the homoeostatic role of FoxO1 in DA system has not been investigated. Here we report that FoxO1 is highly expressed in DA neurons and mice lacking FoxO1 specifically in the DA neurons (FoxO1 KO(DAT)) show markedly increased energy expenditure and interscapular brown adipose tissue (iBAT) thermogenesis accompanied by reduced fat mass and improved glucose/insulin homoeostasis. Moreover, FoxO1 KO(DAT) mice exhibit an increased sucrose preference in concomitance with higher dopamine and norepinephrine levels. Finally, we found that FoxO1 directly targets and negatively regulates tyrosine hydroxylase (TH) expression, the rate-limiting enzyme of the catecholamine synthesis, delineating a mechanism for the KO phenotypes. Collectively, these results suggest that FoxO1 in DA neurons is an important transcriptional factor that directs the coordinated control of energy balance, thermogenesis and glucose homoeostasis.


Figure 3: FoxO1 expression patterns during neuronal differentiation. (a) Endogenous level of miR-9 was determined by quantitative real-time PCR (qRT-PCR) analysis at indicated time points during NSPC differentiation. Pr, proliferating NSPCs. (b) Western blot analysis of FoxO1 and neural markers during NSPC differentiation. Cells were harvested at the indicated time points. GAPDH (glyceraldehyde 3-phosphate dehydrogenase) was used as a loading control. (c–f) The coexpression of FoxO1 with neural markers was determined. FoxO1 staining is shown in green and neural markers, Nestin (c), GFAP (d), βIII-tubulin (e) and DCX (f), are shown in red. Nuclear DAPI (4',6-diamidino-2-phenylindole) staining is shown in blue. (d and e) Filled arrow heads point to cells that are positive for both FoxO1 and markers. (f) Empty arrowheads are for cells that are positive for DCX but do not express FoxO1. Scale bars=50 μm. (g) The percentage of each neural marker-positive cell of the FoxO1-positive cells
Figure 4: Overexpression of FoxO1 inhibits neuronal differentiation. (a) Western blot analysis of βIII-tubulin and DCX expression in Ade-emp- or Ade-FoxO1-infected NSPCs. Cells were differentiated for the indicated time. The overexpression of FoxO1 was also confirmed. (b) Quantitative real-time PCR (qRT-PCR) of Nestin and βIII-tubulin mRNA in Ade-emp- or Ade-FoxO1-infected NSPCs. *P<0.05. (c–e) Immunofluorescence analysis of Nestin (c), GFAP (d) and βIII-tubulin (e) expression after expression of FoxO1-ADA was confirmed. Nuclear DAPI (4',6-diamidino-2-phenylindole) staining is shown in blue. Scale bars=50 μm (c) and 200 μm (d, e). (f) The quantification of Nestin-, GFAP- and βIII-tubulin-positive cell proportions in Ade-emp- or Ade-FoxO1-expressing NSPCs. NS, non-significant
Figure 7: R-9 regulates Notch pathway through FoxO1. (a) CSL-induced luciferase reporter activity was measured in control or miR-9-overexpressed NSPCs. (b) Same experiment as in (a) was performed in wild-type (Ade-emp) or FoxO1-knockout (Ade-Cre) NSPCs. (c) Quantitative real-time PCR (qRT-PCR) analysis of βIII-tubulin and Hes1 mRNA in Ade-emp- or Ade-FoxO1-infected NSPCs, with or without miR-9 overexpression. (d) The FoxO1 occupancy of the Hes1 promoter in NSPCs was determined by chromatin immunoprecipitation (ChIP). (Upper) Predicted FoxO1 binding in the Hes1 promoter. TSS: transcription start site. (Lower) qPCR against two regions in the Hes1 promoter. (e) Western blot analysis of βIII-tubulin and Hes1 expression in Ade-empty- or Ade-Cre-infected FoxO1L/L NSPCs, with (D) or without (V) treatment of 5 μM DAPT. (f) qRT-PCR analysis of βIII-tubulin and Hes1 mRNA in Ade-empty- or Ade-Cre-infected FoxO1L/L NSPCs, with or without DAPT treatment. *P<0.05 and **P<0.01. (g) A graphical presentation of miR-9 and FoxO1 interaction on Notch target gene regulation
Functional regulation of FoxO1 in neural stem cell differentiation

October 2015

·

142 Reads

·

76 Citations

Cell Death and Differentiation

Forkhead transcription factor family O (FoxO) maintains adult stem cell reserves by supporting their long-term proliferative potential. MicroRNAs (miRs) regulate neuronal stem/progenitor cell (NSPC) proliferation and differentiation during neural development by controlling the expression of a specific set of target genes. In the neurogenic subventricular zone, FoxO1 is specifically expressed in NSPCs and is no longer detected during the transition to neuroblast stage, forming an inverse correlation with miR-9 expression. The 3'-untranslated region of FoxO1 contains a conserved target sequence of miR-9 and FoxO1 expression is coordinated in concert with miR-9 during neuronal differentiation. Our study demonstrates that FoxO1 contributes to NSPC fate decision through its cooperation with the Notch signaling pathway.Cell Death and Differentiation advance online publication, 16 October 2015; doi:10.1038/cdd.2015.123.



Forkhead box O transcription factors in chondrocytes regulate endochondral bone formation

July 2015

·

137 Reads

·

24 Citations

The Journal of Steroid Biochemistry and Molecular Biology

The differentiation of embryonic mesenchymal cells into chondrocytes and the subsequent formation of a cartilaginous scaffold that enables the formation of long bones are hallmarks of endochondral ossification. During this process, chondrocytes undergo a remarkable sequence of events involving proliferation, differentiation, hypertrophy and eventually apoptosis. Forkhead Box O (FoxO) transcription factors (TFs) are well-known regulators of such cellular processes. Although FoxO3a was previously shown to be regulated by 1,25-dihydroxyvitamin D3 in osteoblasts, a possible role for this family of TFs in chondrocytes during endochondral ossification remains largely unstudied. By crossing Collagen2-Cre mice with FoxO1(lox/lox);FoxO3a(lox/lox);FoxO4(lox/lox) mice, we generated mice in which the three main FoxO isoforms were deleted in growth plate chondrocytes (chondrocyte triple knock-out; CTKO). Intriguingly, CTKO neonates showed a distinct elongation of the hypertrophic zone of the growth plate. CTKO mice had increased overall body and tail length at eight weeks of age and suffered from severe skeletal deformities at older ages. CTKO chondrocytes displayed decreased expression of genes involved in redox homeostasis. These observations illustrate the importance of FoxO signaling in chondrocytes during endochondral ossification. Copyright © 2015. Published by Elsevier Ltd.


Figure 3 | Absence of FoxOs prevent the induction of critical atrogenes. Quantitative RT-PCR of atrogenes from fed and 24-h starved tibialis anterior of control and FoxO1,3,4 À / À mice. Data are normalized to GAPDH and expressed as fold increase of control-fed animals. n ¼ 4 muscles in each group Values are mean±s.e.m. *Po0.05,**Po0.01. (Student's t-test). 
Figure 4 | Acute inhibition of FoxOs phenocopies the conditional FoxO1,3,4 knockout. (a) Frequency histograms of gastrocnemius muscles showing the distribution of cross-sectional areas (mm 2 ) of inducible muscle-specific FoxO1,3,4 mice after tamoxifen-dependent deletion of FoxO1,3,4 genes (FoxO1,3,4 f/f : black bars and FoxO1,3,4 À / À : magenta bars) in fed (upper panel) and fasted (lower panel) conditions, n ¼ 3, each group. (b) Force measurements preformed in vivo on gastrocnemius muscle showed that acute inhibition of FoxOs in adulthood prevents force drop during fasting. n ¼ 6 muscles in each group. Freq: Frequency. (c) Left, immunoblotting analyses of gastrocnemius homogenates after acute deletion of FoxO1,3,4 À / À and controls. Right, quantification of LC3 lipidation. Data are representative of three independent experiments. (d) Autophagy flux is not increased in FoxO-deficient TA muscles. Inhibition of autophagy-lysosome fusion by colchicine treatment induces accumulation of LC3II band in starved control but not in starved FoxO1,3,4 À / À muscles. Left, immunoblotting analyses of gastrocnemius homogenates. Right, quantification of LC3 lipidation. (e,f) ChIP quantitative RTPCR shows the recruitment of FoxO3 and FoxO1 on the promoters of selected atrophy-related genes. ChIP assays were performed in starved control and FoxO1,3,4 À / À TA muscles. IgG was used as the reference. n ¼ 3 for each group. Data are shown as mean ± s.e.m. Error bars indicate s.e.m. *Po0.05, **Po0.01 (Student's t-test). S1: FoxO binding site 1; S2: FoxO binding site 2. 
Figure 7 | FoxO members are redundant and control a new set of ubiquitin ligases. (a-c) Frequency histograms showing the distribution of cross-sectional areas (mm 2 ) of gastrocnemius muscles from muscle-specific (a) FoxO1 À / À , (b) FoxO3 À / À and (c) FoxO4 À / À mice. Data are shown as mean ± s.e.m. of four muscles each group. *Po0.05, **Po0.01. (d,e) qRT-PCR of the novel ubiquitin ligases MUSA1, Fbxo21/SMART, Fbxo31, Itch from 24 starved (d) or denervated (e) FoxO1,3,4 f/f and FoxO1,3,4 À / À mice. Data are normalized to GAPDH and expressed as fold increase of fed control mice. n ¼ 4 muscles for each group. (f) ChIP qPCR of FoxO3 and (g) FoxO1 on the promoters of MUSA1, Fbxo21/SMART and Itch. IgG was used as the reference. n ¼ 3. (h-j) qRT-PCR of MUSA1, Fbxo21/SMART, Fbxo31, Itch in (h) FoxO1, (i) FoxO3 and (j) FoxO4 knockout mice after 3 days of denervation. Values are normalized to GAPDH and expressed as fold increase of control mice. n ¼ 4 muscles for each group. Data are shown as mean ± s.e.m. Error bars indicate s.e.m. *Po0.05, **Po0.01 (Student's t-test). con control; den, denervated. MUSA1 S1, FoxO-binding site1; MUSA1 S2, FoxO-binding site2; MUSA1 S3, FoxO-binding site3. 
Figure 8 | Smart is a novel ubiquitin ligase required for denervation-dependent atrophy. (a) Co-immunoprecipitation experiment showing that SMART is a F-box protein that forms a SCF complex. C2C12 muscle cell lines were transfected with SMART, Skp1, Cul1 and Roc1 expression plasmids. After 24 h, cells were lysed and immunoprecipitation against FLAG-tag or control IgG was performed. Western blots for the different SCF components are shown. (b) RNAi-mediated knockdown of SMART revealed by quantitative RT-PCR (qRT-PCR). Adult TA muscles were transfected with bicistronic expressing vectors that encode either oligo 4 or scramble and GFP. Two weeks later TA muscles were collected, RNA extracted and endogenous SMART, MUSA1, Atrogin1, MuRF1 and Fbxo31 expression were analysed by qRT-PCR, n ¼ 4. (c) Inhibition of SMART prevents muscle atrophy in denervated muscles. Adult muscle fibres were co-transfected with bicistronic expressing vectors that encode shRNAs against SMART (oligo 4) or scramble and GFP and denervated. Two weeks later cross-sectional area of transfected fibres, identified by GFP fluorescence, was measured. n ¼ 6 muscles for each group. (d) Densitometric quantification of polyubiquitinated proteins in muscle extracts transfected with shRNAi against SMART or scramble. Values are normalized to GAPDH and expressed as fold increase of fed control mice. n ¼ 3 muscles for each group. Data are shown as mean±s.e.m. Error bars indicate s.e.m. *Po0.05, **Po0.01 (Student's t-test). con, control; den, denervated; IB, immunoblotting. 
Regulation of autophagy and the ubiquitin-proteasome system by the FoxO transcriptional network during muscle atrophy

April 2015

·

542 Reads

·

547 Citations

Nature Communications

Stresses like low nutrients, systemic inflammation, cancer or infections provoke a catabolic state characterized by enhanced muscle proteolysis and amino acid release to sustain liver gluconeogenesis and tissue protein synthesis. These conditions activate the family of Forkhead Box (Fox) O transcription factors. Here we report that muscle-specific deletion of FoxO members protects from muscle loss as a result of the role of FoxOs in the induction of autophagy-lysosome and ubiquitin-proteasome systems. Notably, in the setting of low nutrient signalling, we demonstrate that FoxOs are required for Akt activity but not for mTOR signalling. FoxOs control several stress-response pathways such as the unfolded protein response, ROS detoxification, DNA repair and translation. Finally, we identify FoxO-dependent ubiquitin ligases including MUSA1 and a previously uncharacterised ligase termed SMART (Specific of Muscle Atrophy and Regulated by Transcription). Our findings underscore the central function of FoxOs in coordinating a variety of stress-response genes during catabolic conditions.


Citations (63)


... attention as a potential therapeutic strategy for radical cure of cancer [28]; hence, elucidating the mechanisms of CSC population maintenance is crucial for developing anti-tumor therapeutic strategies targeting CSCs [29]. Organelles, such as mitochondria, lysosomes, and endoplasmic reticulum play critical roles in CSC population maintenance [10,30,31]; however, the underlying molecular mechanisms are not completely elucidated. ...

Reference:

p52-ZER6/IGF1R axis maintains cancer stem cell population to promote cancer progression by enhancing pro-survival mitophagy
Mitochondria Define Intestinal Stem Cell Differentiation Downstream of a FOXO/Notch Axis
  • Citing Article
  • November 2020

Cell Metabolism

... To avoid harsh conditions for insulinoma mice, the treatment dose of Car and Car/Hdot was decided as 30 mg/kg of drug (185 mg/kg of Car/H-dot) to be a quarter of the medium lethal dose (LD50) [31], Figure 4a shows color and NIR images of the resected spleen and pancreas with tumors from insulinoma mice on 2 d post-intraperitoneal injection. Strong fluorescence signals of Car/H-dot were observed within the tumor mass (tumor interstitial fluid), similar to our previous results [32]. Presumably, the EPR effect arrowed for quick accumulation and retention of H-dot complexes, resulting in a higher fluorescence signal around the tumor surrounding area, which was retained more than 2 d post-injection. ...

Rapid and Selective Targeting of Heterogeneous Pancreatic Neuroendocrine Tumors

iScience

... Consistently, changes in metabolism as well as mitochondrial genes and proteins have been described in IECs from IBD patients (Mottawea et al., 2016;Rath et al., 2018;Haberman et al., 2019) along with aberrant patterns of LGR5 expression and reduced PC function (Wehkamp et al., 2005;VanDussen et al., 2014;Khaloian et al., 2020). Furthermore, recent data highlight the role of mitochondrial metabolism in deciding on the cellular phenotype and actively determining lineage commitment (Ludikhuize et al., 2019;Khaloian et al., 2020). In line, Paneth cell metaplasia (i.e., occurrence of PCs in the distal colon, where they are physiologically not found) seems to predispose to CRC development (Wada et al., 2005;Pai et al., 2013), and loss of mucin-producing goblet cells is an early event in intestinal inflammation (van der Post et al., 2019;López Cauce et al., 2020). ...

Mitochondria define intestinal stem cell differentiation downstream of a FOXO/Notch axis

... On the molecular level, these metabolic functions are mediated by FoxO1 transcriptional regulation of hypothalamic AgRP/NPY and POMC expression (Kim et al., 2006;Kitamura et al., 2006;Plum et al., 2009;Ren et al., 2012). Furthermore, FoxO1 also modulates the CNS control of energy expenditure by mediating the expression of the steroidogenic factor 1 (SF1) in the ventromedial hypothalamus (VMH) and tyrosine hydroxylase in the dopaminergic neurons (Doan et al., 2016;Kim et al., 2012;Son et al., 2019). In this study, we elucidated the undetermined function of astrocytic FoxO1 in the CNS maintenance of whole-body energy homeostasis. ...

FoxO1 regulates leptin-induced mood behavior by targeting tyrosine hydroxylase
  • Citing Article
  • November 2018

Metabolism

... 1,2 Conversely, mice with OB-specific deletion of either FoxO1 or ATF4, negative regulators of insulin signaling, display the opposite phenotype to INSR OB −/− mice with improved glucose disposal and insulin sensitivity. 3,4 Apart from hepatocytes, OBs are the only cell type in which disruption of insulin signaling hampers glucose metabolism in mice fed a normal chow diet (NCD), 1 further highlighting the importance of the skeleton in regulating glucose homeostasis. ...

FoxO1 expression in osteoblasts regulates glucose homeostasis through regulation of osteocalcin in mice
  • Citing Article
  • March 2010

The Journal of clinical investigation

... Either the KKS or the mesolimbic dopaminergic system are involved in the control of metabolism [27][28][29][30][31]. To investigate the potential impact of B2R deletion in cells expressing TH on energy homeostasis, we conducted an analysis of O 2 consumption and CO 2 production in both female and male mice. ...

FoxO1 in dopaminergic neurons regulates energy homeostasis and targets tyrosine hydroxylase

Nature Communications

... Increased insu- lin/IGF-1 signalling leads to inactivation of FoxO1 and FoxO3a by their phosphorylation-dependent extrusion form the nucleus into the cytoplasm. Both FoxO1 and FoxO3a exert stimulatory effects on FoxP3 expres- sion [40] (Fig. 1). ...

Transcription factors Foxo3a and Foxo1 couple the E3 ligase Cbl-b to the induction of Foxp3 expression in induced regulatory T cells
  • Citing Article
  • May 2010

Journal of Cell Biology (JCB)

Journal of Cell Biology (JCB)

... 32 Nevertheless, these predictions are typically constrained to specific steady-state conditions, and cannot be 33 extrapolated to past or future time points in the cell cycle. Deciphering the kinetic parameters of chromatin 34 accessibility changes could potentially allow gene expression to be predicted over a continuous period 35 of time. However, because the timing and rate of chromatin changes are highly context-dependent and 36 vary strongly between genes, computational methods that can precisely model these kinetic parameters at 37 single-gene resolution are needed. ...

Functional regulation of FoxO1 in neural stem cell differentiation

Cell Death and Differentiation

... We hypothesize that the FOXO3 gene may be involved in muscle formation and developmental processes. Eelen et al. showed that the FOXO3 gene is mainly expressed in skeletal muscle and is associated with muscle atrophy [26]. Muscle atrophy is one of the symptoms of a variety of pathological conditions. ...

Forkhead box O transcription factors in chondrocytes regulate endochondral bone formation
  • Citing Article
  • July 2015

The Journal of Steroid Biochemistry and Molecular Biology

... On the other hand, muscle protein degradation is regulated by signaling pathways involving the forkhead box O (FoxO) and the ubiquitin-proteasome system. In skeletal muscle, there are three isoforms of FoxO (FoxO1, FoxO3a, and FoxO4), and they are predominantly located in the nucleus [40]. When Akt phosphorylates FoxO, it becomes inactivated and moves from the nucleus to the cytoplasm. ...

Regulation of autophagy and the ubiquitin-proteasome system by the FoxO transcriptional network during muscle atrophy

Nature Communications