Adipose Tissue Functions As a Reservoir for Leukemia Stem Cells and Confers Chemo-Resistance

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 845-845 ◽  
Author(s):  
Haobin Ye ◽  
Biniam Adane ◽  
Nabilah Khan ◽  
John M. Ashton ◽  
Marlene Balys ◽  
...  

Abstract Adipose tissue (AT) serves as a storage site for lipids as well as an endocrine organ. In the context of cancer, AT can function to facilitate the progression of tumors. Interestingly, recent studies have shown that AT acts as an extramedullary reservoir of hematopoietic stem cells (HSCs), suggesting the presence of a HSC niche in AT. The fact that leukemia stem cells (LSCs) co-opt the HSC marrow and the supportive effect of AT on cancer cells led us to hypothesize that AT functions as a reservoir for LSCs. To test this hypothesis, we first examined whether there were LSCs residing in AT. Using a murine model of primary blast crisis CML, we found enrichment of phenotypically primitive leukemia cells (PLCs) in the gonadal adipose tissue (GAT) relative to bone marrow, spleen and peripheral blood (Figure 1). The high percentage of PLCs in GAT led us to postulate that PLCs are preferentially reliant on fatty acids as an energy source since GAT was found to be lipolytic in leukemic mice. Indeed, we observed that PLCs had a higher rate of fatty acid oxidation (FAO) compared to lineage+ leukemia cells and their non-leukemic counterparts. Additionally, conditioned medium from adipocytes selectively increased the FAO rate in PLCs suggesting a FAO regulatory role of AT. Further, we showed that CD36, a fatty acid transporter, was highly expressed by a subset of PLCs and selectively regulated FAO in PLCs. Interestingly, CD36+ PLCs were strikingly enriched in GAT. Together, these results suggested that GAT functions as a reservoir for CD36+ PLCs and facilitates use of FAO for energy metabolism. Next we examined the characteristics of CD36+ PLCs. CD36+ PLCs differed metabolically from CD36- PLCs with regard to fatty acid metabolism. Specifically, we found CD36+ PLCs had a higher FAO rate and were more dependent on the transportation function of CD36 for FAO (Figure 2). We also compared the cell cycle status between these two populations and found CD36+ PLCs were more quiescent. Interestingly, both CD36+ and CD36- PLCs contained LSCs and were able to reconstitute the whole leukemic BM when transplanted into recipients. Collectively, these findings indicated that at least two metabolically distinct types of leukemia-initiating cells exist in our blast crisis model, and that the major forms of energy metabolism can differ as a function of anatomical location and expression of CD36. Since front line agents commonly used in cancer generally target actively cycling cells, we speculated that CD36+ PLCs might be more drug resistant due to their increased quiescence. Indeed, we found enrichment of CD36+ PLCs in BM after applying a 5-day chemotherapy regimen to leukemic mice, while CD36- PLCs were not protected. More importantly, we observed that CD36+ PLCs were highly enriched in GAT after chemotherapy suggesting GAT conferred chemo-resistance to resident CD36+ PLCs (Figure 3). Taken together, our observations imply that the heterogeneity found in PLCs is translated into drug sensitivities in different leukemic sub-fractions. Specifically, CD36+ PLCs represent a relatively drug resistant subpopulation and GAT serves as a reservoir for resident CD36+ PLCs. Lastly, we investigated whether these findings in the murine model could be recapitulated in primary human bcCML cells. We found that within the CD34+ subpopulation of primary human bcCML cells, CD36+ subset had a high FAO rate compared to CD36- subset. Furthermore, this CD36+ subset was more quiescent and drug resistant. These data suggest that a similar heterogeneity can be found in primary human bcCML cells based on the expression of CD36. Collectively, our findings suggested that GAT in leukemia mice functions as a reservoir for LSCs and confers chemo-resistance to resident leukemia cells, implying a potential role of GAT in the pathogenesis of leukemia and relative efficacy of therapeutic challenge. Furthermore, our data indicate metabolic heterogeneity within LSC populations, where pathways controlling energy consumption can differ. We propose that metabolic heterogeneity in LSCs may contribute to the challenge in effectively eradicating such cells. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 268-268 ◽  
Author(s):  
Eric A Lee ◽  
Leonard Angka ◽  
Sarah-Grace Rota ◽  
Thomas Hanlon ◽  
Rose Hurren ◽  
...  

Abstract Acute myeloid leukemia (AML) is an aggressive malignant disease characterized by poor patient outcome and suboptimal front-line chemotherapy. To identify novel anti-AML compounds, we performed a high-throughput screen of a natural products library (n=800). This screen was performed against the AML cell line (TEX), which has several properties of leukemia stem cells, the cells responsible for disease pathophysiology and patient relapse. Here, avocatin B was identified as a potent and novel anti-leukemia agent. Avocatin B, at concentrations as high as 20µM, had no effect on normal peripheral blood stem cell viability. In contrast, it induced death of primary AML cells with an EC50 of 1.5-5.0 µM. Selective toxicity towards a functionally defined subset of primitive leukemia cells was also demonstrated. Avocatin B (3µM) reduced clonogenic growth of AML progenitor cells with no effect on clonogenic growth of normal hematopoietic stem cells. Further, treatment of primary AML cells with avocatin B (3µM) diminished their ability to engraft into the bone marrow of pre-conditioned, NOD/SCID mice (t18=6.5; p<0.001). Together, these results confirm that avocatin B is a novel anti-AML agent with selective toxicity toward leukemia and leukemia stem cells. Mechanistically, avocatin B-induced reactive oxygen species (ROS)-dependent leukemia cell apoptosis that was characterized by the release of mitochondrial proteins, cytochrome c and apoptosis inducing factor (AIF). Cytochrome c and AIF were detected in the cytosol of avocatin B treated TEX cells by flow cytometry. Avocatin B-induced apoptosis, as measured by the Annexin V/Propidium iodide assay, DNA fragmentation and PARP cleavage, was abolished in the presence of anti-oxidants confirming the functional importance of ROS. Next, we further evaluated the role of mitochondria in avocatin B-induced apoptosis. First, we generated leukemia cells lacking mitochondria by successive culturing in media containing ethidium bromide. The drastic (>80%) reduction in mitochondria were confirmed by nonyl acridine orange staining and flow cytometry and a near absence of the mitochondria specific proteins ANT and ND1, as measured by Western blotting. Avocatin B’s activity was abolished in leukemia cells lacking mitochondria. Next, using lentiviral knockdown, we generated leukemia cells lacking CPT1, the enzyme that facilitates transport of 16-20 carbon lipids into mitochondria. Avocatin B’s activity was abolished in cells with reduced CPT1 expression (>70% as measured by qPCR analysis). To further confirm the importance of CPT1 in avocatin B-induced death, we chemically inhibited CPT1 with etomoxir. Avocatin B’s activity was blocked in the presence of etomoxir, further demonstrating that avocatin B accumulates in mitochondria. Since avocatin B is a lipid that targets mitochondria and that mitochondria can oxidize fatty acids for energy, we next assessed the impact of avocatin B on fatty acid oxidation, using the Seahorse Bioanalyzer. Avocatin B inhibited leukemia cell fatty acid oxidation (>40% reduction in oxygen consumption at 10µM) and this occurred at a 10-fold less concentration than etomoxir, the standard experimental molecule used to probe this pathway. Further, avocatin B resulted in a 50% reduction in levels of NADPH, an important co-factor generated during fatty acid oxidation that participates in catabolic processes during cell proliferation. These results show that avocatin B accumulates in mitochondria to inhibit fatty acid oxidation and reduce NADPH to result in ROS-mediated leukemia cell apoptosis. This highlights a novel AML-therapeutic strategy by which mitochondria are targeted to impair cellular metabolism leading directly to AML cell death. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Author(s):  
Maria Amaya ◽  
Anagha Inguva ◽  
Shanshan Pei ◽  
Courtney L Jones ◽  
Anna Krug ◽  
...  

AML is characterized by the presence of leukemia stem cells (LSCs), and failure to fully eradicate this population contributes to disease persistence/relapse. Prior studies have characterized metabolic vulnerabilities of LSCs, which demonstrate preferential reliance on oxidative phosphorylation (OXPHOS) for energy metabolism and survival. In the present study, using both genetic and pharmacologic strategies in primary human AML specimens, we show that signal transducer and activator of transcription 3 (STAT3) mediates OXPHOS in LSCs. STAT3 regulates AML-specific expression of MYC, which in turn controls transcription of the neutral amino acid transporter SLC1A5. We show that genetic inhibition of MYC or SLC1A5 acts to phenocopy the impairment of OXPHOS observed with STAT3 inhibition, thereby establishing this axis as a regulatory mechanism linking STAT3 to energy metabolism. Inhibition of SLC1A5 reduces intracellular levels of glutamine, glutathione and multiple TCA metabolites, leading to reduced TCA cycle activity and inhibition of OXPHOS. Based on these findings, we used a novel small molecule STAT3 inhibitor, that binds STAT3 and disrupts STAT3-DNA, to evaluate the biological role of STAT3. We show that STAT3 inhibition selectively leads to cell death in AML stem and progenitor cells derived from newly diagnosed and relapsed patients, while sparing normal hematopoietic cells. Together, these findings establish a STAT3-mediated mechanism that controls energy metabolism and survival in primitive AML cells.


2020 ◽  
Vol 0 (0) ◽  
Author(s):  
Wenjing Liu ◽  
Dahui Li ◽  
Handi Cao ◽  
Haoyun Li ◽  
Yu Wang

AbstractAdipose tissue is an important organ in our body, participating not only in energy metabolism but also immune regulation. It is broadly classified as white (WAT) and brown (BAT) adipose tissues. WAT is highly heterogeneous, composed of adipocytes, various immune, progenitor and stem cells, as well as the stromal vascular populations. The expansion and inflammation of WAT are hallmarks of obesity and play a causal role in the development of metabolic and cardiovascular diseases. The primary event triggering the inflammatory expansion of WAT remains unclear. The present review focuses on the role of adipocyte progenitors (APS), which give rise to specialized adipocytes, in obesity-associated WAT expansion, inflammation and fibrosis.


2021 ◽  
Vol 22 (3) ◽  
pp. 1375
Author(s):  
María Carmen Carceller ◽  
María Isabel Guillén ◽  
María Luisa Gil ◽  
María José Alcaraz

Adipose tissue represents an abundant source of mesenchymal stem cells (MSC) for therapeutic purposes. Previous studies have demonstrated the anti-inflammatory potential of adipose tissue-derived MSC (ASC). Extracellular vesicles (EV) present in the conditioned medium (CM) have been shown to mediate the cytoprotective effects of human ASC secretome. Nevertheless, the role of EV in the anti-inflammatory effects of mouse-derived ASC is not known. The current study has investigated the influence of mouse-derived ASC CM and its fractions on the response of mouse-derived peritoneal macrophages against lipopolysaccharide (LPS). CM and its soluble fraction reduced the release of pro-inflammatory cytokines, adenosine triphosphate and nitric oxide in stimulated cells. They also enhanced the migration of neutrophils or monocytes, in the absence or presence of LPS, respectively, which is likely related to the presence of chemokines, and reduced the phagocytic response. The anti-inflammatory effect of CM may be dependent on the regulation of toll-like receptor 4 expression and nuclear factor-κB activation. Our results demonstrate the anti-inflammatory effects of mouse-derived ASC secretome in mouse-derived peritoneal macrophages stimulated with LPS and show that they are not mediated by EV.


2009 ◽  
Vol 34 (3) ◽  
pp. 315-322 ◽  
Author(s):  
Gregory R. Steinberg

During moderate-intensity exercise, fatty acids are the predominant substrate for working skeletal muscle. The release of fatty acids from adipose tissue stores, combined with the ability of skeletal muscle to actively fine tune the gradient between fatty acid and carbohydrate metabolism, depending on substrate availability and energetic demands, requires a coordinated system of metabolic control. Over the past decade, since the discovery that AMP-activated protein kinase (AMPK) was increased in accordance with exercise intensity, there has been significant interest in the proposed role of this ancient stress-sensing kinase as a critical integrative switch controlling metabolic responses during exercise. In this review, studies examining the role of AMPK as a regulator of fatty acid metabolism in both adipose tissue and skeletal muscle during exercise will be discussed. Exercise induces activation of AMPK in adipocytes and regulates triglyceride hydrolysis and esterfication through phosphorylation of hormone sensitive lipase (HSL) and glycerol-3-phosphate acyl-transferase, respectively. In skeletal muscle, exercise-induced activation of AMPK is associated with increases in fatty acid uptake, phosphorylation of HSL, and increased fatty acid oxidation, which is thought to occur via the acetyl-CoA carboxylase-malony-CoA-CPT-1 signalling axis. Despite the importance of AMPK in regulating fatty acid metabolism under resting conditions, recent evidence from transgenic models of AMPK deficiency suggest that alternative signalling pathways may also be important for the control of fatty acid metabolism during exercise.


2020 ◽  
Author(s):  
E. Matthew Morris ◽  
Roberto D. Noland ◽  
Michael E. Ponte ◽  
Michelle L. Montonye ◽  
Julie A. Christianson ◽  
...  

AbstractCentral integration of peripheral neural signals is one mechanism by which systemic energy homeostasis is regulated. Previous work described increased acute food intake following chemical reduction of hepatic fatty acid oxidation and ATP levels, which was prevented by common hepatic branch vagotomy (HBV). However, possible offsite actions of the chemical compounds confound the precise role of liver energy metabolism. Herein, we used a liver-specific PGC1a heterozygous (LPGC1a) mouse model, with associated reductions in mitochondrial fatty acid oxidation and respiratory capacity, to assess the role of liver energy metabolism in systemic energy homeostasis. LPGC1a male mice have 70% greater high-fat/high-sucrose (HFHS) diet-induced weight gain and 35% greater positive energy balance compared to wildtype (WT) (p<0.05). The greater energy balance was associated with altered feeding behavior and lower activity energy expenditure during HFHS in LPGC1a males. Importantly, no differences in HFHS-induced weight gain or energy metabolism was observed between female WT and LPGC1a mice. WT and LPGC1a mice underwent sham or HBV to assess whether vagal signaling was involved in HFHS-induced weight gain of male LPGC1a mice. HBV increased HFHS-induced weight gain (85%, p<0.05) in male WT, but not LPGC1a mice. As above, sham LPGC1a males gain 70% more weight during short-term HFHS feeding than sham WT (p<0.05). These data demonstrate a sexspecific role of reduced liver energy metabolism in acute diet-induced weight gain, and the need of more nuanced assessment of the role of vagal signaling in short-term diet-induced weight gain.Key Points SummaryReduced liver PGC1a expression results in reduced mitochondrial fatty acid oxidation and respiratory capacity in male mice.Male mice with reduced liver PGC1a expression (LPGC1a) demonstrate greater short-term high-fat/high-sucrose diet-induced weight gain compared to wildtype.Greater positive energy balance during HFHS feeding in male LPGC1a mice is associated with altered food intake patterns and reduced activity energy expenditure.Female LPGC1a mice do not have differences in short-term HFHS-induced body weight gain or energy metabolism compared to wildtype.Disruption of vagal signaling through common hepatic branch vagotomy increases short-term HFHS-induced weight gain in male wildtype mice, but does not alter male LPGC1a weight gain.


2014 ◽  
pp. n/a-n/a ◽  
Author(s):  
Shahnaz Esmaeli ◽  
Abdolamir Allameh ◽  
Masoud Soleimani ◽  
Fatemeh Rahbarizadeh ◽  
Mehdi Frouzandeh-Moghadam

Sign in / Sign up

Export Citation Format

Share Document