Upregulation of Lipid Metabolism Modulators in Myeloma Cells Underlines Their Progression in a Supportive Microenvironment and Linking Metabolic Pathways with Growth Signaling

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 329-329
Author(s):  
Sathisha Upparahallivenkateshaiah ◽  
Khan Sharmin ◽  
Ling Wen ◽  
Rakesh Bam ◽  
Xin Li ◽  
...  

Abstract Abstract 329 Accumulating evidence indicate that cellular metabolism and bi-products also play important roles in signaling associated with tumor cell proliferation, cell cycle, survival and drug resistance. The overall goal of the study was to molecularly characterize MM cells grown in the supportive bone marrow (BM) of clinically relevant SCID-hu or SCID-rab models. MM cells from 22 patients were engrafted in experimental animals. Following establishment of the disease as determined by increased production of circulating human immunoglobulins over a period of 2–4 months, MM cells were isolated from the implanted bones and subjected to global gene expression profile (GEP). Based on stringent criteria (e.g. p<0.05, >2 folds) we identified commonly overexpressed or underexpressed genes in post-engrafted MM cells compared to pre-engrafted cells from the same patients. Among the top upregulated genes we identified several factors associated with lipid metabolism including FABP5 (fatty acid-binding protein 5), SCD (stearoyl CoA desaturase 1), FADS1 (fatty acid desaturase 1) and SLC27A5 (a fatty acid transporter). Clinical GEP data of newly diagnosed patients from Total Therapy program at our institute revealed upregulation of these genes in high risk patients. We further sought to unravel the role of SCD in MM since it has been previously implicated in tumorigenesis and specific inhibitors are being developed for clinical use. SCD (encodes SCD1), is a rate-limiting enzyme responsible for synthesis of monounsaturated fatty acids. We hypothesized that while nutrient unsaturated fatty acids sufficiently satisfy requirement of most normal cells, growing MM cells demand higher content of these lipids for formation of new membrane phospholipids and immediate energy; therefore, inhibiting SCD1 may suppress MM cell survival and proliferation. Small-molecule inhibitor of SCD1 (BioVision) suppressed growth of 5 MM lines dose dependently; 72 hours IC50 ranged between 1μM (p<0.0006) and 2.5 μM (p<0.0001). At 1 μM the SCD1 inhibitor reduced MM cell proliferation by 70±4% (p<0.002) using thymidine incorporation assay and increased number of apoptotic MM cells from 10±1% in control cells to 27±8% in SCD1 inhibitor-treated cells (p<0.03), using annexin V/PI flow cytometry analysis. This inhibitor also disrupted cell cycle progression in MM cell lines as determined by flow cytometry analysis of DNA content. The Akt/mTOR and AMPK pathways, albeit opposing functions, are known central integrators of cellular metabolism and proliferation signaling. SCD1 inhibitor reduced phosphorylated AKT and increased phosphorylated AMPK in MM cells assessed by Western Blot. For in vivo experiments in SCID-rab mice, SCD1 inhibitor was constantly administered (1.25 μg/hour) by osmotic pumps directly connected to the implanted bones that had been engrafted with luciferase-expressing H929 MM cells (6 mice/group). SCD1 inhibitor suppressed MM growth by 60% (p<0.01) assessed by live-animal imaging and measurement of circulating levels of human immunoglobulins in mice sera. These findings suggest that intracellular modulators of lipid metabolism such as SCD1 are induced in MM cells by the supportive BM and mediate signals linking cellular metabolism, survival and proliferation. Disclosures: No relevant conflicts of interest to declare.

2001 ◽  
Vol 29 (2) ◽  
pp. 250-267 ◽  
Author(s):  
R. J. A. Wanders ◽  
P. Vreken ◽  
S. Ferdinandusse ◽  
G. A. Jansen ◽  
H. R. Waterham ◽  
...  

Peroxisomes are subcellular organelles with an indispensable role in cellular metabolism. The importance of peroxisomes for humans is stressed by the existence of a group of genetic diseases in humans in which there is an impairment in one or more peroxisomal functions. Most of these functions have to do with lipid metabolism including the α and β-oxidation of fatty acids. Here we describe the current state of knowledge about peroxisomal fatty acid α- and β-oxidation with particular emphasis on the following: (1) the substrates β-oxidized in peroxisomes; (2) the enzymology of the α- and β-oxidation systems; (3) the permeability properties of the peroxisomal membrane and the role of the different transporters therein; (4) the interaction with other subcellular compartments, including the mitochondria, which are the ultimate site of NADH reoxidation and full degradation of acetyl-CoA to CO2 and water; and (5) the different disorders of peroxisomal α- and β-oxidation.


2006 ◽  
Vol 18 (2) ◽  
pp. 209
Author(s):  
M. A. Ramírez ◽  
E. Pericuesta ◽  
M. Pérez-Crespo ◽  
R. Fernández-González ◽  
P. N. Moreira ◽  
...  

Normally the majority of mammalian cells, including murine embryonic stem (mES) cells, are immersed in a low oxygen environment (hypoxia); however, mES are generally cultured in an atmosphere containing 21% O2 (normoxia). Such conditions may not be the most appropriate for mES propagation. We have tested the effects of hypoxia and culture on either feeder fibroblasts or gelatin substrate on mES cell growth and spontaneous differentiation. Two ES cell lines (R1 129/Sv from the laboratory of A. Nagy and MAR B6D2 F1 generated in our laboratory) were cultured under two different oxygen tensions (5 and 21%), and on two different substrates, 0.1% gelatin or murine embryonic fibroblasts (mEF). Cell cycle, cell proliferation, mRNA expression of pluripotency and differentiation markers, as well as spontaneous differentiation to cardiomyocytes, were monitored. For cell proliferation measurements, mES after 7 days of culture at the different conditions were labeled with 5-(and-6)-carboxyfluorescein diacetate succinimidyl ester, and cultured for up to three more days. Cells were then harvested for flow cytometry analysis every 24 h after labeling (Cell TraceTM CFSE Cell Proliferation Kit; Molecular Probes, Inc., Eugene, OR, USA). For cell cycle analysis, cells grown on mEF under the two different oxygen tensions were fixed after 10 days of culture, and then stained with propidium iodide/Triton-X-100 for flow cytometry analysis (Current Protocols in Cytometry, Chap. 7, 2001). The spontaneous differentiation of embryoid bodies [formed by ES cells in the absence of leukemia inhibitory factor (LIF)] to cardiomyocytes was also monitored. For mRNA expression of pluripotency (Nanog, Oct-3/4, Rex1, GENESIS, FGFR-4, TERF1, Cx43, and GLUT1) and differentiation markers (GATA4, GATA2, AFP, Msx-1, Brachyury, and Myf5), RT-PCR analysis was performed on mES cells from Day 0 to Day 10. Under hypoxia conditions, the proliferation of both types of mES cells was greater than under normoxia, independent of substrate used, and a higher number of apoptotic cells was detected. Moreover, only under normoxia conditions did mES cells lose the expression of pluripotency markers GENESIS and GLUT1. In addition, under lower oxygen tension, the rate of differentiation to beating cardiomyocytes was significantly lower on the feeder layer than that under normoxia (11.9% vs. 28.1%; P = 0.012). The feeder layer supported significantly higher cardiomyocyte formation when compared to 0.1% gelatin at 21% O2 (28.1% vs. 8.3%; P < 0.001). Our results show that normoxia may not be the most appropriate condition for mES cell propagation and that hypoxic culture may be necessary to maintain full pluripotency of mES cells.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1614-1614
Author(s):  
Cristina Panaroni ◽  
Keertik Fulzele ◽  
Tomoaki Mori ◽  
Rie Nakamoto-Matsubara ◽  
Allison Maebius ◽  
...  

Abstract Obesity-induced increases in bone marrow adipocyte (BMAd) numbers and volume are associated with an increased risk of multiple myeloma (MM). We analyzed gene expression from previously published public datasets and found that 11 out of 47 genes associated with fatty acid (FA) metabolism showed increasing trend from MGUS (monoclonal gammopathy of undetermined significance), MM, to plasma cell leukemia. These genes included ACC1, the first and rate-limiting step of de novo fatty acid biosynthesis and ECHS1, the second step of the mitochondrial fatty acid beta-oxidation pathway. We have previously shown that adipocytes support the growth of MM cells. However, the molecular mechanisms of interaction between MM cells and BMAd remain largely unknown. Here, we hypothesize that BMAd support MM cells through metabolic reprogramming. Here we sought to identify the molecular pathways involved in MM/ BMAd interaction. BM aspirates of MGUS, smoldering MM (SMM), and newly diagnosed MM (NDMM) patients were used to isolate fat-enriched BM fraction and BM stromal cells (BMSCs). Murine BMSC cell-line OP9, murine MM cell-line 5TGM1, and human MM cell lines MM.1S and OPM2 were obtained from ATCC or provided by collaborators and cultured as their respective standard procedures. In-vitro adipogenesis was induced in OP9 cells or BMSCs by supplementing media with dexamethasone, indomethacin, insulin, and IBMX. MM cells were co-cultured directly with pre- or mature adipocytes. Cell proliferation was assessed using CyQUANT NF Cell Proliferation Assay. Lipolysis was assessed by High Sensitivity Lipolysis Assay Kit (Sigma). Lipid uptake in MM cells was assessed by flow-cytometry analysis of the incorporation of fluorescent 12- or 16-carbon long-chain fatty acids BODIPY-FL-C12 and BODIPY-FL-C16, respectively, or LipidTox labelled FA from co-cultured adipocytes. In-vivo effects of excess FA on MM cell growth were assessed using a plasmacytoma model in CB17 SCID mice. In-vitro co-culture revealed that BMSC-derived adipocytes (Ad) from MGUS, SMM and NDMM donors increased the proliferation of MM.1S MM cells significantly. Similarly, mature murine OP9 Ad cells also increased the proliferation of 5TGM1 murine MM cells. Interestingly, co-cultures showed dramatic decrease in LipidTox-stained lipid-droplet size distribution, suggesting increased lipolysis in Ad. In the process of lipolysis, various lipase enzymes hydrolyze stored triglycerides into free fatty acids (FFA) and glycerol. Co-culture of 5TGM1 or OPM2 cells with OP9 mature Ad increased glycerol secretion in the conditioned media by more than 3-fold indicating that MM induce lipolysis in Ad. Consequently, BMAd from MGUS, SMM, and NDMM patients showed increased expression of genes responsible for lipolysis (NR1H3) and increased FA desaturation (SCD1, FASD2). Although MM cells lacked intracellular lipid storage, OPM2 and 5TGM1 MM cells rapidly took up BODIPY-C12 and -C16 FAs. The FA secreted from Ad were directly taken up by MM cells as shown by transfer of LipidTox-labeled lipids from OP9 Ad to unstained 5TGM1 or OPM2 MM cells as assessed by flow cytometry. The addition of Acipomox, a small-molecule inhibitor of lipolysis, decreased LipidTox signal in MM cells compared to untreated OP9 cells. FA are primarily transported into cells through FATP (1-6) or CD36 receptors. Bioinformatic analysis of public database showed that FATP1 and FATP4 were highly expressed in 21 human MM cell lines. Indeed, MM cells from NDMM patients expressed high levels of FATP1 and FATP4. The uptake of BODIPY-C12 and -C16 by 5TGM1 or OPM2 MM cells was significantly reduced in the presence of Lipofermata, a pharmacological small-molecule inhibitor of FATP. Lipidomic analysis of BM aspirates from MM patients showed altered expression of various FA, including arachidonic acid (AA). Low doses of AA (0.125 - 2 µM) increased the proliferation and viability of MM cells whereas high doses (25- 100µM) dramatically decreased it, indicating a bimodal cellular effect of AA. Peritumoral AA treatment in a plasmacytoma model in CB17 SCID mice using MM.1S cells dramatically decreased tumor volume along with the markers of proliferation. In summary, we show that MM cells induce lipolysis in BMAd and that the released FFA are then taken up by MM cells through FATPs. Inhibition of either BMAd lipolysis or FFA transporter into MM cells could be a potential novel strategy to prevent MM progression. Disclosures Fulzele: Constellation Pharma: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company. Raje: Celgene, Amgen, Bluebird Bio, Janssen, Caribou, and BMS: Other.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 10-11
Author(s):  
Connor S Murphy ◽  
Heather Fairfield ◽  
Mariah Farrell ◽  
Victoria DeMambro ◽  
Samantha Costa ◽  
...  

Multiple myeloma (MM) is defined by the clonal expansion of malignant plasma cells in the bone marrow (BM) and has a 5-year survival rate of 50% (Siegel el al. 2018, Cancer J. Clin.). MM remains incurable due to the development of resistance to current chemotherapies; therefore, it is paramount to investigate novel treatments and the mechanisms of drug resistance in MM cells. Interestingly, obesity correlates with increased incidence of MM and high body mass index correlates with a poor treatment response (Marinac et al. 2019, JNCI Cancer Spectr, Groß et al. 2017, Oncotarget). Obesity is a major risk factor for many cancers, however, given the complexity of obesity, there are an array of mechanisms by which obesity may support tumor cells. Studies of obesity and MM are mainly at the epidemiological level and have not extensively explored the mechanism of this relationship. Therefore, there is a critical need to understand how obesity contributes to support cancers such as MM. Possible mechanisms may be through the increased availability of free fatty acids or through other factors that are found in obese patients. We hypothesize that lipid metabolism contributes to obesity-linked cancers such as MM. Recently, changes in lipid metabolism have been shown to support the proliferation, migration and the development of drug resistance in other blood cancers such as acute myeloid leukemia (Tabe et al. 2017, Cancer Res, Tabe et al. 2018, Sci. Reports) and solid tumors such as breast (Wang et al. 2017, JCI Insights) and prostate (Mitra et al. 2017, BMC Cancer) cancer. However, the role of lipid metabolism in MM cells has been understudied. Therefore, we hypothesized that genes within the Hallmark Fatty Acid Metabolism gene set (https://www.gsea-msigdb.org) would be differentially expressed between healthy patients and those with MM. We mined the clinical data (GSE6477, Chng et al. 2007, Cancer Res.) and found that transcripts of an enzyme critical for lipid metabolism, acyl-CoA synthetase long chain member 1 (ACSL1), was significantly downregulated (Figure 1A, Log2(Fold Change)=-2.33, adjusted p value=1.64*10-5, false discovery rate) in patients with newly diagnosed MM relative to normal plasma cells. Therefore, we hypothesized that ACSL1 may act as a tumor suppressor in MM. In order to test the role of the ACSL family as tumor suppressors, we treated human (MM1.S, OPM2 and RPMI-8226) and mouse myeloma (5TGM1) cell lines with an inhibitor (Triacsin C, TriC) of four of the five human acyl-CoA synthetase long chain family members (ACSL1,3,4 and 5). Contrary to our hypothesis, TriC treatment significantly decreased MM cell proliferation (Figure 1B, p&lt;0.0001, One-way ANOVA Tukey's multiple comparisons test is used throughout unless otherwise noted), increased apoptosis (Figure 1C, p&lt;0.001) and caused G0 arrest (Figure1D, p&lt;0.0001) in a dose-dependent manner. Motivated to understand if TriC's toxicity was due to changes in metabolic dynamics, MM1.S cells were treated with 1 μM TriC for 30 minutes and subjected to a metabolic flux assay (Seahorse XF, Agilent). TriC treatment significantly reduced ATP-dependent respiration from fatty acid oxidation (FAO) (Figure 1F, p&lt;0.0001 Student's t-test) and increased proton leak (p&lt;0.0001). Taken together, our data demonstrate that TriC-mediated ACSL inhibition in MM cells decreases proliferation, induces G0 arrest, apoptosis and decreases FAO-dependent respiration and mitochondrial function. It is unclear what ACSL family member is responsible for the phenotype we report here. To address these questions, future studies will focus on genetically targeting individual ACSL family members and characterizing the lipidomic profile of MM ACSL mutants. Our data also suggests that fatty acids are used as an energy source, therefore we will explore how FAO contributes to MM cell proliferation and survival. Disclosures No relevant conflicts of interest to declare.


1961 ◽  
Vol 200 (4) ◽  
pp. 847-850 ◽  
Author(s):  
Judith K. Patkin ◽  
E. J. Masoro

Cold acclimation is known to alter hepatic lipid metabolism. Liver slices from cold-acclimated rats have a greatly depressed capacity to synthesize long-chain fatty acids from acctate-1-C14. Since adipose tissue is the major site of lipogenic activity in the intact animal, its fatty acid synthetic capacity was studied. In contrast to the liver, it was found that adipose tissue from the cold-acclimated rat synthesized three to six times as much long-chain fatty acids per milligram of tissue protein as the adipose tissue from the control rat living at 25°C. Evidence is presented indicating that adipose tissue from cold-acclimated and control rats esterify long-chain fatty acids at the same rate. The ability of adipose tissue to oxidize palmitic acid to CO2 was found to be unaltered by cold acclimation. The fate of the large amount of fatty acid synthesized in the adipose tissue of cold-acclimated rats is discussed.


2015 ◽  
Vol 101 (4) ◽  
pp. 870-878 ◽  
Author(s):  
Mette Bohl ◽  
Ann Bjørnshave ◽  
Kia V Rasmussen ◽  
Anne Grethe Schioldan ◽  
Bashar Amer ◽  
...  

ABSTRACT Background: Abdominal obesity and exaggerated postprandial lipemia are independent risk factors for cardiovascular disease (CVD) and mortality, and both are affected by dietary behavior. Objective: We investigated whether dietary supplementation with whey protein and medium-chain saturated fatty acids (MC-SFAs) improved postprandial lipid metabolism in humans with abdominal obesity. Design: We conducted a 12-wk, randomized, double-blinded, diet intervention study. Sixty-three adults were randomly allocated to one of 4 diets in a 2 × 2 factorial design. Participants consumed 60 g milk protein (whey or casein) and 63 g milk fat (with high or low MC-SFA content) daily. Before and after the intervention, a high-fat meal test was performed. We measured changes from baseline in fasting and postprandial triacylglycerol, apolipoprotein B-48 (apoB-48; reflecting chylomicrons of intestinal origin), free fatty acids (FFAs), insulin, glucose, glucagon, glucagon-like peptide 1 (GLP-1), and gastric inhibitory polypeptide (GIP). Furthermore, changes in the expression of adipose tissue genes involved in lipid metabolism were investigated. Two-factor ANOVA was used to examine the difference between protein types and fatty acid compositions, as well as any interaction between the two. Results: Fifty-two participants completed the study. We found that the postprandial apoB-48 response decreased significantly after whey compared with casein (P = 0.025) independently of fatty acid composition. Furthermore, supplementation with casein resulted in a significant increase in the postprandial GLP-1 response compared with whey (P = 0.003). We found no difference in postprandial triacylglycerol, FFA, insulin, glucose, glucagon, or GIP related to protein type or MC-SFA content. We observed no interaction between milk protein and milk fat on postprandial lipemia. Conclusion: We found that a whey protein supplement decreased the postprandial chylomicron response compared with casein in persons with abdominal obesity, thereby indicating a beneficial impact on CVD risk. This trial was registered at clinicaltrials.gov as NCT01472666.


Author(s):  
Daniel Lunn ◽  
James G Wallis ◽  
John Browse

Abstract A central goal of green chemistry is to produce industrially useful fatty acids in oilseed crops. Although genes encoding suitable fatty acid-modifying enzymes are available from more than a dozen wild species, progress has been limited because expression of these enzymes in transgenic plants produces only low yields of the desired products. For example, fatty acid hydroxylase 12 (FAH12) from castor (Ricinus communis) produces only 17% hydroxy fatty acids (HFAs) when expressed in Arabidopsis (Arabidopsis thaliana), compared to 90% HFA in castor seeds. The transgenic plants also have reduced oil content and seed vigor. Here, we review experiments that have provided for steady increases HFA accumulation and oil content. This research has led to exciting new discoveries of enzymes and regulatory processes in the pathways of both seed oil synthesis and lipid metabolism in other parts of the plant. Recent investigations have revealed that HFA-accumulating seeds are unable to rapidly mobilize HFA- containing triacylglycerol (TAG) storage lipid after germination to provide carbon and energy for seedling development, resulting in reduced seedling establishment. These findings present a new opportunity to investigate a different, key area of lipid metabolism - the pathways of TAG lipolysis and β-oxidation in germinating seedlings.


2018 ◽  
Vol 19 (11) ◽  
pp. 3325 ◽  
Author(s):  
Lucille Stuani ◽  
Fabien Riols ◽  
Pierre Millard ◽  
Marie Sabatier ◽  
Aurélie Batut ◽  
...  

Background: In Acute Myeloid Leukemia (AML), a complete response to chemotherapy is usually obtained after conventional chemotherapy but overall patient survival is poor due to highly frequent relapses. As opposed to chronic myeloid leukemia, B lymphoma or multiple myeloma, AML is one of the rare malignant hemopathies the therapy of which has not significantly improved during the past 30 years despite intense research efforts. One promising approach is to determine metabolic dependencies in AML cells. Moreover, two key metabolic enzymes, isocitrate dehydrogenases (IDH1/2), are mutated in more than 15% of AML patient, reinforcing the interest in studying metabolic reprogramming, in particular in this subgroup of patients. Methods: Using a multi-omics approach combining proteomics, lipidomics, and isotopic profiling of [U-13C] glucose and [U-13C] glutamine cultures with more classical biochemical analyses, we studied the impact of the IDH1 R132H mutation in AML cells on lipid biosynthesis. Results: Global proteomic and lipidomic approaches showed a dysregulation of lipid metabolism, especially an increase of phosphatidylinositol, sphingolipids (especially few species of ceramide, sphingosine, and sphinganine), free cholesterol and monounsaturated fatty acids in IDH1 mutant cells. Isotopic profiling of fatty acids revealed that higher lipid anabolism in IDH1 mutant cells corroborated with an increase in lipogenesis fluxes. Conclusions: This integrative approach was efficient to gain insight into metabolism and dynamics of lipid species in leukemic cells. Therefore, we have determined that lipid anabolism is strongly reprogrammed in IDH1 mutant AML cells with a crucial dysregulation of fatty acid metabolism and fluxes, both being mediated by 2-HG (2-Hydroxyglutarate) production.


2019 ◽  
Author(s):  
Mohammad Aziz ◽  
Saeed Al Mahri ◽  
Amal Alghamdi ◽  
Maaged AlAkiel ◽  
Monira Al Aujan ◽  
...  

Abstract Background Colorectal cancer is a worldwide problem which has been associated with changes in diet and lifestyle pattern. As a result of colonic fermentation of dietary fibres, short chain free fatty acids are generated which activate Free Fatty Acid Receptors 2 and 3 (FFAR2 and FFAR3). FFAR2 and FFAR3 genes are abundantly expressed in colonic epithelium and play an important role in the metabolic homeostasis of colonic epithelial cells. Earlier studies point to the involvement of FFAR2 in colorectal carcinogenesis. Methods Transcriptome analysis console was used to analyse microarray data from patients and cell lines. We employed shRNA mediated down regulation of FFAR2 and FFAR3 genes which was assessed using qRT-PCR. Assays for glucose uptake and cAMP generation was done along with immunofluorescence studies. For measuring cell proliferation, we employed real time electrical impedance based assay available from xCelligence. Results Microarray data analysis of colorectal cancer patient samples showed a significant down regulation of FFAR2 gene expression. This prompted us to study the FFAR2 in colorectal cancer. Since, FFAR3 shares significant structural and functional homology with FFAR2, we knocked down both these receptors in colorectal cancer cell line HCT 116. These modified cell lines exhibited higher proliferation rate and were found to have increased glucose uptake as well as increased level of GLUT1. Since, FFAR2 and FFAR3 signal through G protein subunit (Gαi), knockdown of these receptors was associated with increased cAMP. Inhibition of PKA did not alter the growth and proliferation of these cells indicating a mechanism independent of cAMP/PKA pathway. Conclusion: Our results suggest role of FFAR2/FFAR3 genes in increased proliferation of colon cancer cells via enhanced glucose uptake and exclude the role of protein kinase A mediated cAMP signalling. Alternate pathways could be involved that would ultimately result in increased cell proliferation as a result of down regulated FFAR2/FFAR3 genes. This study paves the way to understand the mechanism of action of short chain free fatty acid receptors in colorectal cancer.


Sign in / Sign up

Export Citation Format

Share Document