Metabolic Cross Talk Between Foxo3 and mTOR Is Essential for Hematopoietic Stem Cell Function

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 856-856 ◽  
Author(s):  
Pauline Rimmele ◽  
Valentina d'Escamard ◽  
Kocabas Fatih ◽  
Chengcheng Zhang ◽  
Hesham A. Sadek ◽  
...  

Abstract Abstract 856 Unbalanced accumulation of reactive oxygen species (ROS) compromises self-renewal of hematopoietic stem cells (HSC). Nonetheless, survival of HSC in the hypoxic niche requires mitochondrial ROS-mediated activation of hypoxic response and significant metabolic adaptation. Hypoxia favors glycolysis to mitochondrial oxidative phosphorylation for energy production. In agreement with this, increased mitochondrial function results in defective maintenance of HSC as observed in Tsc1−/− mice through overactivation of mammalian target of rapamycin (mTOR) signaling pathway. Mechanisms that regulate the strict coordination of mitochondrial function with upstream metabolic pathways that is required for HSC maintenance and balancing ROS remain largely unknown. Foxo3 Forkhead transcription factor is a strong candidate for coordinating metabolic pathways in HSC. Evidence from our laboratory and others has identified Foxo3 as a critical regulator of HSC quiescence and a key modulator of oxidative stress in HSC. To address whether Foxo3 has a more global metabolic control of HSC activity, we investigated the mitochondrial function in Foxo3 mutant HSC. To achieve this, we measured ATP content and oxygen consumption, two major defining mitochondrial parameters, using ATP Bioluminescence Assay and Oxygen Biosensor analysis respectively in freshly isolated Lin−Sca-1+cKit+(LSK) CD34−Flk2− bone marrow cells isolated from wild type and Foxo3−/− mice. We showed that loss of Foxo3 leads to significant mitochondrial defects in HSC as indicated by a strong decrease in both ATP content and oxygen consumption. In agreement with this, the glycolytic flux in Foxo3 mutant HSC, as analyzed by 13C lactate production using gas chromatography-mass spectrometry, was increased indicating a shift in the ATP production from mitochondria to the cytosolic glycolysis in Foxo3 mutant HSC. In addition, mitochondrial mass and membrane potential, that is generated during mitochondrial oxidative phosphorylation for energy production, were assessed by flow cytometry in WT and Foxo3−/− LSK using mitotracker green and JC-1 probes respectively. Loss of Foxo3 increased both mitochondrial mass and membrane potential, likely reflecting a compensatory mechanism to the defective mitochondria in Foxo3−/− HSC. These anomalies may partly contribute, in addition to the known defective ROS detoxification, to the increased ROS levels previously observed in Foxo3−/− HSC. Importantly, the mitochondrial dysfunction was not due to the abnormal increase of ROS observed in Foxo3−/− HSC since in vivo treatment of Foxo3−/− mice with ROS scavenger N-Acetyl-Cysteine (NAC) for two weeks, did not revert the increased mitochondrial membrane potential. Altogether these results strongly suggest that Foxo3 is critical for the regulation of mitochondrial function in HSC. mTOR signaling controls major cellular metabolic processes and is critical for the regulation of mitochondrial function in HSC. We have previously found that mTOR signaling is amplified by a redox-mediated mechanism in Foxo3−/− hematopoietic progenitors (Yalcin et al., 2010). Thus, we asked whether mTOR signaling is involved in the Foxo3 regulation of mitochondria function in HSC. Interestingly, in vivo treatment of Foxo3−/− mice for two weeks with rapamycin, a specific inhibitor of mTOR complex 1 (mTORC1) activity, as measured by the decrease in phosphorylation of ribosomal protein S6 in HSC, mitigates the increased mitochondrial membrane potential and normalizes ROS levels in Foxo3−/− HSC, suggesting that Foxo3 regulation of mitochondria is mediated by mTOR signaling in HSC. Notably, rapamycin treatment rescued partially Foxo3−/− HSC pool and function as measured by the number and frequency of LSK in Foxo3−/− mice, as well as their long-term repopulation ability measured by the capacity of CD48−CD150+LSK cells to repopulate the hematopoietic compartment in lethally irradiated recipient mice within 8 weeks. Taken together, our findings reveal a new function for Foxo3 in the control of mitochondria in HSC and support a model in which mitochondria is key to the maintenance of HSC. We propose that a Foxo3-mTOR signaling node partly controls mitochondrial function in HSC. These findings are likely to have an important impact on our understanding of the metabolic regulation of hematopoietic and leukemic stem cells and may be of therapeutic value. Disclosures: No relevant conflicts of interest to declare.

2020 ◽  
Vol 21 (10) ◽  
pp. 3592
Author(s):  
Dilip Bhargava Sreerangaraja Urs ◽  
Wen-Han Wu ◽  
Katerina Komrskova ◽  
Pavla Postlerova ◽  
Yung-Feng Lin ◽  
...  

Ovarian follicle steroidogenesis associated with embryo quality results in a successful pregnancy. Each follicle consists of an oocyte surrounded by granulosa cells, which secrete several steroid and peptide hormones. Follicles harvested from women who conceived after assisted reproductive therapy (ART) had significantly higher estradiol levels in follicular fluids than the follicles from women who failed to conceive after ART. The higher follicular estradiol levels correlate well with successful fertilization following ART. Mitochondria are the central sites for steroid hormone biosynthesis. The first and rate-limiting step in the biosynthesis of steroid hormones occurs in the mitochondria of granulosa cells. In the present study, we hypothesized that the mitochondria in granulosa cells are critical for maintaining oocyte quality and fertility capacity. This study aims to clarify the relationship between mitochondrial function and granulosa cell steroidogenesis, and the relationship between hormone levels and fertility capacity. Sera, follicular fluids and granulosa cells were obtained from individuals undergoing IVF-ET treatment. The oocyte numbers, oocyte quality, fertilization rate, and pregnancy rate were also recorded. The patients who provided the granulosa cells were further classified into four groups: endometriosis, ovarian endometrioma, endometriosis without ovarian endometrioma, and polycystic ovary syndrome (PCOS); patients with other female factor infertility and male factor infertility were used as controls. We measured the levels of estradiol (E2) by radioimmunoassay. Concurrently, we analyzed the mitochondrial mass and membrane potential, and apoptosis by flow cytometry using nonyl acridine orange, TMRE, Annexin V-FITC and PI. Mitochondrial morphology was visualized by transfection with pLV-mitoDsRed. In addition, we assessed the protein levels of steroidogenic enzymes, steroidogenic acute regulatory protein (StAR) and 3β-hydroxysteroid dehydrogenase (3β-HSD) by Western blot. The results showed significantly decreased serum E2 and follicular E2 levels, and decreased IVF outcomes, in the patients with endometriosis. Reduced mitochondrial mass and decreased mitochondrial membrane potential were correlated with lower E2. Furthermore, a significant decrease in StAR and 3β-HSD was found in patients with ovarian endometrioma. The enzyme levels of StAR and 3β-HSD were highly correlated with E2 levels. Finally, elevated cumulus cell apoptosis was found in the patient group with ovarian endometrioma and PCOS. In conclusion, mitochondrial dysfunction of human granulosa cells may contribute to the decline of steroidogenesis, decreased fertilization rate, oocyte maturation rate, and oocyte quality, and it can ultimately jeopardize fertility.


Author(s):  
Mukul Girotra ◽  
Anne-Christine Thierry ◽  
Alexandre Harari ◽  
George Coukos ◽  
Olaia Naveiras ◽  
...  

2019 ◽  
Vol 2019 ◽  
pp. 1-10 ◽  
Author(s):  
Luena Papa ◽  
Mansour Djedaini ◽  
Ronald Hoffman

Quiescent and self-renewing hematopoietic stem cells (HSCs) rely on glycolysis rather than on mitochondrial oxidative phosphorylation (OxPHOS) for energy production. HSC reliance on glycolysis is considered an adaptation to the hypoxic environment of the bone marrow (BM) and reflects the low energetic demands of HSCs. Metabolic rewiring from glycolysis to mitochondrial-based energy generation accompanies HSC differentiation and lineage commitment. Recent evidence, however, highlights that alterations in mitochondrial metabolism and activity are not simply passive consequences but active drivers of HSC fate decisions. Modulation of mitochondrial activity and metabolism is therefore critical for maintaining the self-renewal potential of primitive HSCs and might be beneficial for ex vivo expansion of transplantable HSCs. In this review, we emphasize recent advances in the emerging role of mitochondria in hematopoiesis, cellular reprograming, and HSC fate decisions.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4787-4787
Author(s):  
Xia Xiao ◽  
Hongmei Luo ◽  
Amanda C. LaRue ◽  
Bradley A. Schulte ◽  
Yong Wang

Abstract Abstract 4787 The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that regulates protein synthesis, gene transcription, cell growth and cell proliferation. Previous studies have demonstrated that abnormalities in the Pten-mTOR pathway may contribute to the development of leukemia and lead to premature exhaustion of hematopoietic stem cells (HSCs). These findings suggest a role for mTOR in the regulation of HSC self-renewal and cell transformation. The aim of this study was to investigate the involvement of mTOR signaling in the survival and proliferation of HSCs and hematopoietic progenitor cells (HPCs). Immunofluorescent staining with antibodies against phosphorylated mTOR and S6 kinase revealed that Thrombopoietin (Tpo) activates mTOR in cultured mouse bone marrow (BM) lineage negative cells, suggesting that Tpo may promote the survival and proliferation of HSCs/HPCs via activation of the mTOR signaling pathway. Further studies revealed that treatment with an mTOR specific small molecule inhibitor (Ku-69734) significantly suppressed the colony-forming ability of HPCs as evidenced by a dose-dependent decrease in the production of CFU-GM, BFU-E and CFU-GEMM. We also examined the clonogenic function of HSCs using cobblestone-area forming cell (CAFC) assays and found that Ku-69734 treatment markedly reduced the number of CAFCs in long-term BM culture. Moreover, immunophenotyping and flow cytometric analyses showed that inhibition of mTOR induced apoptosis primarily in HSCs and to a lesser degree in HPCs, indicating that mTOR inhibition may suppress the clonogenic function of HSCs and HPCs via the induction of apoptosis. Together, these data demonstrate that mTOR signaling is required for the survival and proliferation of HSCs and HPCs. Given that many mTOR inhibitors are currently in clinical trials for the treatment of cancers, our findings provide the rationale to further evaluate the potential toxicity of mTOR inhibitors in HSCs/HPCs in vivo. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 2013 ◽  
pp. 1-7 ◽  
Author(s):  
Haizhen Mo ◽  
Ying Chen ◽  
Liyong Huang ◽  
Hao Zhang ◽  
Juxiang Li ◽  
...  

Tea polyphenols are of great benefit to the treatment of several neurodegenerative diseases. In order to explore the neuroprotective effects of tea polyphenols and their potential mechanisms, an establishedin vivosubarachnoid hemorrhage (SAH) model was used and alterations of mitochondrial function, ATP content, and cytochromec(cytc) in cerebral cortex were detected. This study showed that the alteration of mitochondrial membrane potential was an early event in SAH progression. The trend of ATP production was similar to that of mitochondrial membrane potential, indicating that the lower the mitochondrial membrane potential, lesser the ATP produced. Due to mitochondrial dysfunction, more cytcwas released in the SAH group. Interestingly, the preadministration of tea polyphenols significantly rescued the mitochondrial membrane potential to basal level, as well as the ATP content and the cytclevel in the brain cortex 12 h after SAH. After pretreatment with tea polyphenols, the neurological outcome was also improved. The results provide strong evidence that tea polyphenols enhance neuroprotective effects by inhibiting polarization of mitochondrial membrane potential, increasing ATP content, and blocking cytcrelease.


2010 ◽  
Vol 299 (1) ◽  
pp. E14-E22 ◽  
Author(s):  
Eva Corpeleijn ◽  
Nina P. Hessvik ◽  
Siril S. Bakke ◽  
Klaus Levin ◽  
Ellen E. Blaak ◽  
...  

Obesity and insulin resistance are related to both enlarged intramyocellular triacylglycerol stores and accumulation of lipid intermediates. We investigated how lipid overflow can change the oxidation of intramyocellular lipids (ICLOX) and intramyocellular lipid storage (ICL). These experiments were extended by comparing these processes in primary cultured myotubes established from healthy lean and obese type 2 diabetic (T2D) individuals, two extremes in a range of metabolic phenotypes. ICLs were prelabeled for 2 days with 100 μM [14C]oleic acid (OA). ICLOX was studied using a 14CO2 trapping system and measured under various conditions of extracellular OA (5 or 100 μM) and glucose (0.1 or 5.0 mM) and the absence or presence of mitochondrial uncoupling [carbonyl cyanide p-trifluoromethoxyphenylhydrazone (FCCP)]. First, increased extracellular OA availability (5 vs. 100 μM) reduced ICLOX by 37%. No differences in total lipolysis were observed between low and high OA availability. Uncoupling with FCCP restored ICLOX to basal levels during high OA availability. Mitochondrial mass was positively related to ICLOX, but only in myotubes from lean individuals. In all, a lower mitochondrial mass and lower ICLOX were related to a higher cell-associated OA accumulation. Second, myotubes established from obese T2D individuals showed reduced ICLOX. ICLOX remained lower during uncoupling ( P < 0.001), even with comparable mitochondrial mass, suggesting decreased mitochondrial function. Furthermore, the variation in ICLOX in vitro was significantly related to the in vivo fasting respiratory quotient of all subjects ( P < 0.02). In conclusion, the rate of ICLOX is dependent on the availability of extracellular fatty acids and mitochondrial function rather than mitochondrial mass.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5257-5257
Author(s):  
Wenbin Zhong ◽  
Vesa Olkkonen ◽  
Xu Bing ◽  
Biying Zhu ◽  
Guoping Li ◽  
...  

Abstract Acute myelogenous leukemia (AML) is one of the deadliest hematological malignancies and there is at present no efficient strategy to defeat it. New detailed insight into AML leukemia stem cells (LSCs) survival will facilitate the identification of targets for the development of new therapeutic approaches. Recent work has provided evidence that LSCs are defective in their ability to employ glycolysis, but are highly reliant on oxidative phosphorylation, and the maintenance of mitochondrial function is essential for LSCs survival. It is increasingly clear that Ca2+ released constitutively from endoplasmic reticulum (ER) is taken up by mitochondria to sustain optimal bioenergetics and cell survival. Here we report three striking findings: 1) oxysterol-binding protein (OSBP)-related protein 4 (ORP4L) is expressed in LSCs but not in normal hematopoietic stem cells (HSCs). 2) ORP4L is essential for LSC bioenergetics; It forms a complex with PLCβ3 and IP3 receptor 1 (ITPR1) to control Ca2+ release from the ER and subsequent cytosolic and mitochondrial parallel Ca2+ spike oscillations that sustain pyruvate dehydrogenase (PDH) activation and oxidative phosphorylation. 3) ORP4L inhibition eradicates LSCs in vitro and in vivo through impairment of Ca2+-dependent bioenergetics. These results suggest a novel role of ORP4L in governing Ca2+ release to sustain mitochondrial function and survival of LSCs and identify ORP4L as a putative new oncoprotein and therapeutic target for LSCs elimination. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 6 (15) ◽  
pp. eaax5150
Author(s):  
Zachary J. Milstone ◽  
Sherin Saheera ◽  
Lauren M. Bourke ◽  
Tomer Shpilka ◽  
Cole M. Haynes ◽  
...  

Cryptic transcription occurs widely across the eukaryotic genome; however, its regulation during vertebrate development is not understood. Here, we show that two class I histone deacetylases, Hdac1 and Hdac2, silence cryptic transcription to promote mitochondrial function in developing murine hearts. Mice lacking Hdac1 and Hdac2 in heart exhibit defective developmental switch from anaerobic to mitochondrial oxidative phosphorylation (OXPHOS), severe defects in mitochondrial mass, mitochondrial function, and complete embryonic lethality. Hdac1/Hdac2 promotes the transition to OXPHOS by enforcing transcriptional fidelity of metabolic gene programs. Mechanistically, Hdac1/Hdac2 deacetylates histone residues including H3K23, H3K14, and H4K16 to suppress cryptic transcriptional initiation within the coding regions of actively transcribed metabolic genes. Thus, Hdac1/2-mediated epigenetic silencing of cryptic transcription is essential for mitochondrial function during early vertebrate development.


2018 ◽  
Vol 19 (11) ◽  
pp. 3483
Author(s):  
James Phelan ◽  
Finbar MacCarthy ◽  
Dermot O’Toole ◽  
Narayanasamy Ravi ◽  
John Reynolds ◽  
...  

Barrett’s esophagus and esophageal cancer lack prognostic markers that allow the tailoring of personalized medicine and biomarkers with potential to provide insight into treatment response. This study aims to characterize mitochondrial function across the metaplasia-dysplasia-adenocarcinoma disease sequence in Barrett’s esophagus and examines the functional effect of manipulating mitochondrial genes. Mitochondrial genes of interest were validated in in vitro cell lines across the metaplasia (QH), dysplasia (GO) and adenocarcinoma (OE33) sequence and in in vivo patient tissue samples. These genes were subsequently knocked down in QH and OE33 cells and the functional effect of siRNA-induced knockdown on reactive oxygen species production, mitochondrial mass, mitochondrial membrane potential and cellular metabolism was investigated. Three global mitochondrial genes (BAK1, FIS1 and SFN) were differentially altered across the in vivo Barrett’s disease sequence. We also demonstrate that knockdown of BAK1, FIS1 and SFN in vitro resulted in significant alterations in mitochondrial membrane potential; however, no differences in reactive oxygen species or mitochondrial mass were observed. Furthermore, knockdown of these genes in esophageal adenocarcinoma cells significantly altered cellular metabolism. In conclusion, we found that differential expression of BAK1, FIS1, and SFN were altered across the Barrett’s disease sequence and manipulation of these genes elicited significant effects on mitochondrial membrane potential.


2020 ◽  
Author(s):  
Silpa Gampala ◽  
Fenil Shah ◽  
Xiaoyu Lu ◽  
Hye-ran Moon ◽  
George Sandusky ◽  
...  

Abstract Background: Pancreatic cancer is a complex disease with a desmoplastic stroma, extreme hypoxia, and inherent resistance to therapy. Understanding the signaling and adaptive response of such an aggressive cancer is key to making advances in therapeutic efficacy. Redox factor-1 (Ref-1), a redox signaling protein, regulates the DNA binding activity of several transcription factors (TFs), including HIF-1α, STAT3 and NFκB . The conversion of these TFs from an oxidized to reduced state lead to enhancement of their DNA binding. In our previously published work, knockdown of Ref-1 under normoxia resulted in altered gene expression patterns on pathways including EIF2, protein kinase A, and mTOR. In this study, single cell RNA sequencing (scRNA-seq) and proteomics were used to explore the effects of Ref-1 on metabolic pathways under hypoxia.Methods: scRNA-seq comparing pancreatic cancer cells expressing less than 20% of the Ref-1 protein was analyzed using left truncated mixture Gaussian model and validated using proteomics and qRT-PCR. The impact of knocking down Ref-1 under hypoxia was dramatic on the mitochondria and metabolic pathways. Ref-1’s role in mitochondrial function was confirmed using mitochondrial function assays and qRT-PCR. Further, the effect of Ref-1 redox function inhibition against pancreatic cancer metabolism was assayed using 3D co-culture in vitro and xenograft studies in vivo and compared to Devimistat, a drug that disrupts mitochondrial metabolism.Results: Distinct transcriptional variation in central metabolism, cell cycle, apoptosis, immune response, and genes downstream of a series of signaling pathways and transcriptional regulatory factors were identified in Ref-1 knockdown vs Scrambled control from the scRNA-seq data. Mitochondrial DEG subsets downregulated with Ref-1 knockdown were significantly reduced following Ref-1 redox inhibition and more dramatically in combination with Devimistat in vitro. Mitochondrial function assays demonstrated that Ref-1 knockdown and Ref-1 redox signaling inhibition decreased utilization of TCA cycle substrates and slowed the growth of pancreatic cancer co-culture spheroids. In vivo xenograft studies demonstrated that tumor reduction was potent with Ref-1 redox inhibitor similar to Devimistat.Conclusion: Ref-1 redox signaling inhibition conclusively alters cancer cell metabolism by causing TCA cycle dysfunction while also reducing the pancreatic tumor growth in vitro as well as in vivo.


Sign in / Sign up

Export Citation Format

Share Document