scholarly journals Remodeling of whole-body lipid metabolism and a diabetic-like phenotype caused by loss of CDK1 and hepatocyte division

eLife ◽  
2020 ◽  
Vol 9 ◽  
Author(s):  
Jin Rong Ow ◽  
Matias J Cadez ◽  
Gözde Zafer ◽  
Juat Chin Foo ◽  
Hong Yu Li ◽  
...  

Cell cycle progression and lipid metabolism are well-coordinated processes required for proper cell proliferation. In liver diseases that arise from dysregulated lipid metabolism, proliferation is diminished. To study the outcome of CDK1 loss and blocked hepatocyte proliferation on lipid metabolism and the consequent impact on whole-body physiology, we performed lipidomics, metabolomics, and RNA-seq analyses on a mouse model. We observed reduced triacylglycerides in liver of young mice, caused by oxidative stress that activated FOXO1 to promote expression of Pnpla2/ATGL. Additionally, we discovered that hepatocytes displayed malfunctioning β-oxidation, reflected by increased acylcarnitines (ACs) and reduced β-hydroxybutyrate. This led to elevated plasma free fatty acids (FFAs), which were transported to the adipose tissue for storage and triggered greater insulin secretion. Upon aging, chronic hyperinsulinemia resulted in insulin resistance and hepatic steatosis through activation of LXR. Here, we demonstrate that loss of hepatocyte proliferation is not only an outcome but also possibly a causative factor for liver pathology.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3946-3946
Author(s):  
Liping Li ◽  
Katharina Hayer ◽  
Lingling Xian ◽  
Li Luo ◽  
Leslie Cope ◽  
...  

Introduction: Acute B-cell lymphoblastic leukemia (B-ALL) is the most common form of childhood leukemia and the leading cause of death in children with cancer. While therapy is often curative, about 10-15% of children will relapse with recurrent disease and abysmal outcomes. Actionable mechanisms that mediate relapse remain largely unknown. The gene encoding the High Mobility Group A1(HMGA1) chromatin regulator is overexpressed in diverse malignancies where high levels portend poor outcomes. In murine models, we discovered thatHmga1 overexpression is sufficient for clonal expansion and progression to aggressive acute lymphoid leukemia (Cancer Res 2008,68:10121, 2018,78:1890; Nature Comm 2017,8:15008). Further, HMGA1 is overexpressed in pediatric B-ALL (pB-ALL) blasts with highest levels in children who relapse early compared to those who achieve chronic remissions. Together, these findings suggest that HMGA1 is required for leukemogenesis and may foster relapse in B-ALL. We therefore sought to: 1) test the hypothesis that HMGA1 is a key epigenetic regulator required for leukemogenesis and relapse in pB-ALL, and, 2) elucidate targetable mechanisms mediated by HMGA1 in leukemogenesis. Methods: We silenced HMGA1 via lentiviral delivery of short hairpin RNAs targeting 2 different sequences in cell lines derived from relapsed pB-ALL (REH, 697). REH cells harbor the TEL-AML1 fusion; 697 cells express BCL2, BCL3, and cMYC. Next, we assessed leukemogenic phenotypes in vitro (proliferation, cell cycle progression, apoptosis, and clonogenicity) and leukemogenesis invivo. To dissect molecular mechanisms underlying HMGA1, we performed RNA-Seq and applied in silico pathway analysis. Results: There is abundant HMGA1 mRNA and protein in both pB-ALL cell lines and HMGA1 was effectively silenced by short hairpin RNA. Further, silencing HMGA1 dramatically halts proliferation in both cell lines, leading to a decrease in cells in S phase with a concurrent increase in G0/S1. Apoptosis also increased by 5-10% after HMGA1 silencing based on flow cytometry for Annexin V. In colony forming assays, silencing HMGA1 impaired clonogenicity in both pB-ALL cell lines. To assess HMGA1 function in leukemogenesis in vivo, we implanted control pB-ALL cells (transduced with control lentivirus) or those with HMGA1 silencing via tail vein injection into immunosuppressed mice (NOD/SCID/IL2 receptor γ). All mice receiving control REH cells succumbed to leukemia with a median survival of only 29 days. At the time of death, mice had marked splenomegaly along with leukemic cells circulating in the peripheral blood and infiltrating both the spleen and bone marrow. In contrast, mice injected with REH cells with HMGA1 silencing survived for >40 days (P<0.001) and had a significant decrease in tumor burden in the peripheral blood, spleen, and bone marrow. Similar results were obtained with 697 cells, although this model was more fulminant with control mice surviving for a median of only 17 days. To determine whether the leukemic blasts found in mice injected with ALL cells after HMGA1 silencing represented a clone that expanded because it escaped HMGA1 silencing, we assessed HMGA1 levels and found that cells capable of establishing leukemia had high HMGA1 expression, with levels similar to those observed in control cells without HMGA1 silencing. RNA-Seq analyses from REH and 697 cell lines with and without HMGA1 silencing revealed that HMGA1 up-regulates transcriptional networks involved in RAS/MAPK/ERK signaling while repressing the IDH1 metabolic gene, the latter of which functions in DNA and histone methylation. Studies are currently underway to identify effective agents to target HMGA1 pathways. Conclusions: Silencing HMGA1 dramatically disrupts leukemogenic phenotypes in vitro and prevents the development of leukemia in mice. Mechanistically, RNA-Seq analyses revealed that HMGA amplifies transcriptional networks involved cell cycle progression and epigenetic modifications. Our findings highlight the critical role for HMGA1 as a molecular switch required for leukemic transformation in pB-ALL and a rational therapeutic target that may be particularly relevant for relapsed B-ALL. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 8 (12) ◽  
pp. 316
Author(s):  
Xiaoxiao Gao ◽  
Xiaolei Yao ◽  
Xiaodan Li ◽  
Yaxu Liang ◽  
Zifei Liu ◽  
...  

The uterus, as part of the female reproductive tract, is essential for embryo survival and in the maintenance of multiple pregnancies in domestic animals. This study was conducted to investigate the effects of WNT6 on Hu sheep endometrial epithelial cells (EECs) and uterine glands (UGs) in Hu sheep, with high prolificacy rates. In the present study, Hu sheep with different fecundity, over three consecutive pregnancies, were divided into two groups: high prolificacy rate group (HP, litter size = 3) and low prolificacy rate group (LP, litter size = 1). A comparative analysis of the endometrial morphology was performed by immunofluorescence. RNA-seq was used to analyze the gene’s expression in endometrium of HP and LP Hu sheep, providing a candidate gene, which was investigated in EECs and organoid culture. Firstly, higher density of UGs was found in the HP Hu sheep groups (p < 0.05). The RNA-seq data revealed the importance of the WNT signaling pathway and WNT6 gene in Hu sheep endometrium. Functionally, WNT6 could promote the cell cycle progression of EECs via WNT/β-catenin signal and enhance UGs organogenesis. Taken together, WNT6 is a crucial regulator for sheep endometrial development; this finding may offer a new insight into understanding the regulatory mechanism of sheep prolificacy.


2020 ◽  
Author(s):  
Shawn A. Hallett ◽  
Wanida Ono ◽  
Yuki Matsushita ◽  
Naoko Sakagami ◽  
Koji Mizuhashi ◽  
...  

AbstractChondrocytes in the resting zone of the postnatal growth plate are characterized by slow cell cycle progression, and encompass a population of parathyroid hormone-related protein (PTHrP)-expressing skeletal stem cells that contribute to the formation of columnar chondrocytes. However, how these chondrocytes are maintained in the resting zone remains undefined. We undertook a genetic pulse-chase approach to isolate slow cycling, label-retaining chondrocytes (LRCs) from the growth plate using a chondrocyte-specific doxycycline-controllable Tet-Off system regulating expression of histone 2B-linked GFP. Comparative RNA-seq analysis identified significant enrichment of inhibitors and activators for Wnt/β-catenin signaling in LRCs and non-LRCs, respectively. Activation of Wnt/β-catenin signaling in PTHrP+ resting chondrocytes using Pthrp-creER and Apc-floxed allele impaired their ability to form columnar chondrocytes. Therefore, slow-cycling chondrocytes are maintained in a canonical Wnt-inhibitory environment within the resting zone, unraveling a novel mechanism regulating maintenance and differentiation of PTHrP+ skeletal stem cells of the postnatal growth plate.


2020 ◽  
Author(s):  
Cara E. Shields ◽  
Sindhu Potlapalli ◽  
Selma M. Cuya-Smith ◽  
Sarah K. Chappell ◽  
Dongdong Chen ◽  
...  

ABSTRACTRhabdomyosarcoma (RMS) is an aggressive pediatric soft tissue sarcoma that continues to present significant challenges to pediatric oncology. There are two major subtypes of pediatric rhabdomyosarcoma, alveolar and embryonal. Alveolar rhabdomyosarcomas are characterized by the presence of a PAX-FOXO1 fusion protein and termed fusion-positive (FP-RMS); embryonal rhabdomyosarcomas (ERMS) lack these fusions and are termed fusion-negative (FN-RMS).Fusion-positive rhabdomyosarcoma (FP-RMS) harbors PAX-FOXO1 fusion proteins and has a worse overall outcome compared to FN-RMS, underscoring the critical need to identify novel targets for this disease. While fusion proteins remain challenging therapeutic targets, recent studies have begun to reveal the key intersection of PAX-FOXO1 fusion proteins with the malignant epigenome, suggesting that epigenetic proteins may serve as novel targets in FP-RMS. Here, we investigate the contribution of the epigenetic regulator BMI1 to FP-RMS.We examined RNA-seq tumor datasets and determined that BMI1 is robustly expressed in FP-RMS tumors, patient derived xenografts (PDXs), and cell lines. We depleted BMI1 using RNA interference and find that this leads to a marked decrease in cell growth. Next, we used two BMI1 inhibitors, PTC-209 and PTC-028, and showed that BMI1 inhibition decreases cell cycle progression and increases apoptosis in FP-RMS cell lines. In the in vivo setting, targeting BMI1 leads to decreased tumor growth. Mechanistically, we observe that BMI1 inhibition activates the tumor suppressive Hippo pathway. Collectively, these results identify BMI1 as a novel therapeutic vulnerability in FP-RMS and provide a foundation for further investigation of BMI1 in both FP-RMS and additional sarcoma histotypes.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2679-2679
Author(s):  
Mona M.Hosseini ◽  
Hsin-Yun Lin ◽  
Gabby Dewson ◽  
Ruthey Vivier ◽  
Anupriya Agarwal

Background: Whole-genome sequencing and expression studies have revealed significant heterogeneity in the molecular abnormalities driving AML. Selective inhibitors have been developed for many of the pathways influenced by these genetic alterations, but successful translation of these agents into the clinic is limited by both disease heterogeneity and drug resistance. Targeting of inflammatory pathways to block leukemia progression and eliminate leukemic clones is an emerging concept in AML therapy. We have shown that elevated levels of pro-inflammatory cytokine, interleukin-1 (IL-1), in AML microenvironment enhances the growth of leukemic progenitors in variety of genetic subtypes while inhibiting the normal progenitors' growth. To reveal molecular mechanisms underlying such paradoxical effect, we performed RNA-seq analysis on AML and healthy progenitors post IL-1 stimulation. We found myristoylated alanine-rich C-kinase substrate (MARCKS) is one of the most differentially expressed genes in AML progenitors compared to healthy progenitors. MARCKS is a major substrate of protein kinase C, and plays a crucial role in cell survival, migration, and cell cycle progression. Increased MARCKS expression promotes metastasis in solid tumors and inhibiting its activation is being proposed as a therapeutic strategy. However, its role in AML has not yet been investigated. Here, we show a crucial role of MARCKS activation in IL-1-mediated leukemia progression. Method and Results: Using the RNA-seq gene expression data of 451 primary AML patient samples (Tyner et al., Nature 2018), we tested the correlation of MARCKS with IL1R1 receptor expression in AML primary samples and found it to be positively correlated (r = 0.45, p < 0.0001). The correlation was regardless of sex, age, and mutation status. Using q-PCR and western blot analysis, we showed that MARCKS expression, protein level, and its activation (phosphorylation) are elevated in AML samples at basal level and after IL-1 stimulation when compared to the healthy progenitors (~3 fold change). These results validated our transcriptome data and suggested an important role for MARCKS in IL-1-mediated AML progression. To identify the functional significance of MARCKS in AML, we used two independent doxycycline inducible shRNAs to knockdown MARCKS in AML cell lines (MOLM-14 and THP-1). Our data show that MARCKS depletion in AML cells reduces the cell viability overtime to 40%, cell growth to 4 fold, and colony formation ability to 2 fold. Mechanistically, the knockdown of MARCKS in AML cells decreased SKP2 and increased p27 protein levels, suggesting MARCKS regulates cell cycle progression in these cells. We xenografted MOLM-14 cells expressing MARCKS shRNA into NSG mice by tail vein injections and induced the knockdown in vivo by feeding mice doxycycline containing chow. The bone marrow and spleen cells were analyzed by flow cytometry for human and mouse cell markers approximately 3 weeks post-treatment. We observed that the knockdown of MARCKS decreased the leukemia burden in xenograft model as observed by ~80% reduction in human leukemia cells in the bone marrow, ~40% reduction in human leukemia cells in spleen, and ~50% reduction in spleen size compared to the controls, suggesting MARCKS has a critical role in leukemia progression. Conclusion: MARCKS is over-expressed and -activated in various AML genetic subtypes. IL-1 stimulation of AML progenitors increases MARCKS phosphorylation. MARCKS promotes AML progression by increasing the cellular growth, survival, and cell cycle progression of leukemic cells. These results suggest that MARCKS may serve as marker for IL-1 mediated inflammatory stress and offers a route for new targeted therapy. Disclosures No relevant conflicts of interest to declare.


2014 ◽  
Vol 28 (S1) ◽  
Author(s):  
Maria Chondronikola ◽  
Craig Porter ◽  
Nicholas Hurren ◽  
Tony Chao ◽  
Christina Yfanti ◽  
...  

2020 ◽  
Vol 117 (29) ◽  
pp. 17177-17186 ◽  
Author(s):  
Heng Wu ◽  
Yitzhak Reizel ◽  
Yue J. Wang ◽  
Jessica L. Lapiro ◽  
Betsy T. Kren ◽  
...  

Hepatocyte nuclear factor 4α (HNF4α) is a master regulator of liver function and a tumor suppressor in hepatocellular carcinoma (HCC). In this study, we explore the reciprocal negative regulation of HNF4α and cyclin D1, a key cell cycle protein in the liver. Transcriptomic analysis of cultured hepatocyte and HCC cells found that cyclin D1 knockdown induced the expression of a large network of HNF4α-regulated genes. Chromatin immunoprecipitation-sequencing (ChIP-seq) demonstrated that cyclin D1 inhibits the binding of HNF4α to thousands of targets in the liver, thereby diminishing the expression of associated genes that regulate diverse metabolic activities. Conversely, acute HNF4α deletion in the liver induces cyclin D1 and hepatocyte cell cycle progression; concurrent cyclin D1 ablation blocked this proliferation, suggesting that HNF4α maintains proliferative quiescence in the liver, at least, in part, via repression of cyclin D1. Acute cyclin D1 deletion in the regenerating liver markedly inhibited hepatocyte proliferation after partial hepatectomy, confirming its pivotal role in cell cycle progression in this in vivo model, and enhanced the expression of HNF4α target proteins. Hepatocyte cyclin D1 gene ablation caused markedly increased postprandial liver glycogen levels (in a HNF4α-dependent fashion), indicating that the cyclin D1-HNF4α axis regulates glucose metabolism in response to feeding. In AML12 hepatocytes, cyclin D1 depletion led to increased glucose uptake, which was negated if HNF4α was depleted simultaneously, and markedly elevated glycogen synthesis. To summarize, mutual repression by cyclin D1 and HNF4α coordinately controls the cell cycle machinery and metabolism in the liver.


2007 ◽  
Vol 292 (1) ◽  
pp. E107-E116 ◽  
Author(s):  
Anne L. Friedlander ◽  
Kevin A. Jacobs ◽  
Jill A. Fattor ◽  
Michael A. Horning ◽  
Todd A. Hagobian ◽  
...  

To evaluate the contribution of working muscle to whole body lipid oxidation, we examined the effects of exercise intensity and endurance training (9 wk, 5 days/wk, 1 h, 75% V̇o2 peak) on whole body and leg free fatty acid (FFA) kinetics in eight male subjects (26 ± 1 yr, means ± SE). Two pretraining trials [45 and 65% V̇o2 max (45UT, 65UT)] and two posttraining trials [65% of pretraining V̇o2 peak (ABT), and 65% of posttraining V̇o2 peak (RLT)] were performed using [1-13C]palmitate infusion and femoral arteriovenous sampling. Training increased V̇o2 peak by 15% (45.2 ± 1.2 to 52.0 ± 1.8 ml·kg−1·min−1, P < 0.05). Muscle FFA fractional extraction was lower during exercise (EX) compared with rest regardless of workload or training status (≈20 vs. 48%, P < 0.05). Two-leg net FFA balance increased from net release at rest (≈−36 μmol/min) to net uptake during EX for 45UT (179 ± 75), ABT (236 ± 63), and RLT (136 ± 110) ( P < 0.05), but not 65UT (51 ± 127). Leg FFA tracer measured uptake was higher during EX than rest for all trials and greater during posttraining in RLT (716 ± 173 μmol/min) compared with pretraining (45UT 450 ± 80, 65UT 461 ± 72, P < 0.05). Leg muscle lipid oxidation increased with training in ABT (730 ± 163 μmol/min) vs. 65UT (187 ± 94, P < 0.05). Leg muscle lipid oxidation represented ∼62 and 30% of whole body lipid oxidation at lower and higher relative intensities, respectively. In summary, training can increase working muscle tracer measured FFA uptake and lipid oxidation for a given power output, but both before and after training the association between whole body and leg lipid metabolism is reduced as exercise intensity increases.


2014 ◽  
Vol 307 (11) ◽  
pp. G1073-G1087 ◽  
Author(s):  
Bryan C. Tackett ◽  
Hongdan Sun ◽  
Yu Mei ◽  
Janielle P. Maynard ◽  
Sayuri Cheruvu ◽  
...  

Extracellular nucleotides via activation of P2 purinergic receptors influence hepatocyte proliferation and liver regeneration in response to 70% partial hepatectomy (PH). Adult hepatocytes express multiple P2Y (G protein-coupled) and P2X (ligand-gated ion channels) purinergic receptor subtypes. However, the identity of key receptor subtype(s) important for efficient hepatocyte proliferation in regenerating livers remains unknown. To evaluate the impact of P2Y2 purinergic receptor-mediated signaling on hepatocyte proliferation in regenerating livers, wild-type (WT) and P2Y2 purinergic receptor knockout (P2Y2−/−) mice were subjected to 70% PH. Liver tissues were analyzed for activation of early events critical for hepatocyte priming and subsequent cell cycle progression. Our findings suggest that early activation of p42/44 ERK MAPK (5 min), early growth response-1 (Egr-1) and activator protein-1 (AP-1) DNA-binding activity (30 min), and subsequent hepatocyte proliferation (24–72 h) in response to 70% PH were impaired in P2Y2−/− mice. Interestingly, early induction of cytokines (TNF-α, IL-6) and cytokine-mediated signaling (NF-κB, STAT-3) were intact in P2Y2−/− remnant livers, uncovering the importance of cytokine-independent and nucleotide-dependent early priming events critical for subsequent hepatocyte proliferation in regenerating livers. Hepatocytes isolated from the WT and P2Y2−/− mice were treated with ATP or ATPγS for 5–120 min and 12–24 h. Extracellular ATP alone, via activation of P2Y2 purinergic receptors, was sufficient to induce ERK phosphorylation, Egr-1 protein expression, and key cyclins and cell cycle progression of hepatocytes in vitro. Collectively, these findings highlight the functional significance of P2Y2 purinergic receptor activation for efficient hepatocyte priming and proliferation in response to PH.


Sign in / Sign up

Export Citation Format

Share Document