Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yunsheng Li is active.

Publication


Featured researches published by Yunsheng Li.


Experimental and Clinical Endocrinology & Diabetes | 2015

Glucose- and Triglyceride-lowering Dietary Penta-O-galloyl-α-D-Glucose Reduces Expression of PPARγ and C/EBPα, Induces p21-Mediated G1 Phase Cell Cycle Arrest, and Inhibits Adipogenesis in 3T3-L1 Preadipocytes.

X. Liu; Ahmed Malki; Yanyang Cao; Yunsheng Li; Yanrong Qian; Xuan Wang; Xiaozhuo Chen

Plant polyphenols, such as hydrolysable tannins, are present in the human diet and known to exhibit anti-diabetic and anti-obesity activity. We previously reported that the representative hydrolysable tannin compound α-penta-galloyl-glucose (α-PGG) is a small molecule insulin mimetic that, like insulin, binds to insulin receptor (IR) and activates the IR-Akt-GLUT4 signaling pathway to trigger glucose transport and reduce blood glucose levels in db/db and ob/ob diabetic mice. However, its effects on adipogenesis and lipid metabolism were not known. In this study, high fat diet (HFD)-induced diabetic and obese mice were treated with α-PGG to determine its effects on blood glucose and triglycerides. 3T3-L1 preadipocytes were used as a cell model for identifying the anti-adipogenic activity of α-PGG at molecular and cellular levels as a first step in elucidating the mechanism of action of the compound. In vivo, oral administration of α-PGG significantly reduced levels of blood glucose, triglyceride, and insulin in HFD-induced diabetic/obese mice (P<0.05). In vitro, α-PGG inhibited the differentiation of 3T3-L1 preadipocytes into mature adipocytes. α-PGG suppressed the expression of positive adipogenic factors PPARγ C/EBPα and mTOR and augmented the negative adipogenic factor Pref-1. Furthermore, α-PGG induced upregulation of p21 and G1 phase cell cycle arrest. In contrast, adipogenic signaling pathways mediated by insulin, the cAMP response element binding protein (CREB) and glucocorticoid receptor (GR), were not inhibited. RNAi knockdown of p21 led to a 4-fold increase in triglyceride level in 3T3-L1 preadipocytes treated with MDI and α-PGG compared to regular preadipocytes. These results indicate, for the first time, that α-PGG is blood triglyceride- and glucose-lowering in HFD-induced obese and diabetic mice. It selectively inhibited some but not all major adipogenic pathways as well as the mTOR-p21-mediated cell cycle regulatory pathway. It is very likely that these apparently diverse but coordinated activities together inhibited adipogenesis. These results expand our knowledge on how PGG works in adipocytes and further confirm that α-PGG functions as an orally-deliverable natural insulin mimetic with adipogenetic modulatory functions.


Journal of Molecular Endocrinology | 2013

Orally efficacious novel small molecule 6-chloro-6-deoxy-1,2,3,4-tetra-O-galloyl-α-d-glucopyranose selectively and potently stimulates insulin receptor and alleviates diabetes

Yanyan Cao; Yunsheng Li; Jaekyung Kim; Yulin Ren; Klaus Himmeldirk; Yi Liu; Yanrong Qian; Fengming Liu; Xiaozhuo Chen

Type 2 diabetes (T2D) has become an epidemic worldwide while T1D remains a great medical challenge. Insulin receptor (IR) signaling activators could alleviate hyperglycemia, reduce the burden on the pancreas, and contribute to prevention and treatment of both types of diabetes. Previously, we reported the synthesis and identification of a natural antidiabetic compound α-penta-galloyl-glucose (α-PGG). Subsequent studies led to the identification of an α-P6GG derivative, 6-chloro-6-deoxy-1,2,3,4-tetra-O-galloyl-α-D-glucopyranose (6Cl-TGQ). Here, we report that 6Cl-TGQ not only induced rapid and long-lasting glucose uptake comparable to insulin in adipocytes but also reduced high blood glucose levels to near normal and significantly decreased plasma insulin levels and improved glucose tolerance performance in high-fat diet-induced T2D mice when administered orally at 5 mg/kg once every other day. Moreover, a single gavage of 6Cl-TGQ at 10 mg/kg induced rapid and sharp decline of blood glucose in streptozotocin-induced T1D mice. Our studies further indicated that 6Cl-TGQ activated IR signaling in cell models and insulin-responsive tissues of mice. 6Cl-TGQ-induced Akt phosphorylation was completely blocked by IR and PI3K inhibitors, while the induced glucose uptake was blocked by the same compounds and a Glut4 inhibitor. Receptor binding studies indicated that 6Cl-TGQ bound to IR with a higher affinity than α-PGG. Importantly, 6Cl-TGQ, unlike insulin, selectively induced phosphorylation of IR without activating IGF1R or its signaling and did not increase cancer cell proliferation. These results indicate that 6Cl-TGQ is a potent orally efficacious compound with low carcinogenic potential and may contribute to the prevention and treatment of T1D and T2D.


Molecular Cancer Research | 2016

Extracellular ATP a New Player in Cancer Metabolism: NSCLC Cells Internalize ATP in vitro and in vivo Using Multiple Endocytic Mechanisms.

Yanrong Qian; Xuan Wang; Yunsheng Li; Yanyang Cao; Xiaozhuo Chen

Intratumoral extracellular ATP concentrations are 1000 times higher than those in normal tissues of the same cell origin. However, whether or not cancer cells use the abundant extracellular ATP was unknown until we recently reported that cancer cells internalize ATP. The internalized ATP was found to substantially increase intracellular ATP concentration and promote cell proliferation and drug resistance in cancer cells. Here, using a nonhydrolyzable fluorescent ATP (NHF-ATP), radioactive and regular ATP, coupled with high and low molecular weight dextrans as endocytosis tracers and fluorescence microscopy and ATP assays, cultured human NSCLC A549 and H1299 cells as well as A549 tumor xenografts were found to internalize extracellular ATP at concentrations within the reported intratumoral extracellular ATP concentration range. In addition to macropinocytosis, both clathrin- and caveolae-mediated endocytosis significantly contribute to the ATP internalization, which led to an approximately 30% (within 45 minutes) or more than 50% (within 4 hours) increase in intracellular ATP levels after ATP incubation. This increase could not be accounted for by either purinergic receptor signaling or increased intracellular ATP synthesis rates in the ATP-treated cancer cells. These new findings significantly deepen our understanding of the Warburg effect by shedding light on how cancer cells in tumors, which are heterogeneous for oxygen and nutrition supplies, take up extracellular ATP and use the internalized ATP to perform multiple previously unrecognized functions of biological importance. They strongly suggest the existence of ATP sharing among cancer and stromal cells in tumors and simultaneously identify multiple new anticancer targets. Implications: Extracellular ATP is taken up by human lung cancer cells and tumors via macropinocytosis and other endocytic processes to supplement their extra energy needs for cancer growth, survival, and drug resistance, thus providing novel targets for future cancer therapy. Mol Cancer Res; 14(11); 1087–96. ©2016 AACR.


Oncotarget | 2017

Extracellular ATP, as an energy and phosphorylating molecule, induces different types of drug resistances in cancer cells through ATP internalization and intracellular ATP level increase

Xuan Wang; Yunsheng Li; Yanrong Qian; Yanyang Cao; Pratik Shriwas; Haiyun Zhang; Xiaozhuo Chen

Cancer cells are able to uptake extracellular ATP (eATP) via macropinocytosis to elevate intracellular ATP (iATP) levels, enhancing their survival in drug treatment. However, the involved drug resistance mechanisms are unknown. Here we investigated the roles of eATP as either an energy or a phosphorylating molecule in general drug resistance mediated by ATP internalization and iATP elevation. We report that eATP increased iATP levels and promoted drug resistance to various tyrosine kinase inhibitors (TKIs) and chemo-drugs in human cancer cell lines of five cancer types. In A549 lung cancer cells, the resistance was downregulated by macropinocytosis inhibition or siRNA knockdown of PAK1, an essential macropinocytosis enzyme. The elevated iATP upregulated the efflux activity of ABC transporters in A549 and SK-Hep-1 cells as well as phosphorylation of PDGFRα and proteins in the PDGFR-mediated Akt-mTOR and Raf-MEK signaling pathways in A549 cells. Similar phosphorylation upregulations were found in A549 tumors. These results demonstrate that eATP induces different types of drug resistance by eATP internalization and iATP elevation, implicating the ATP-rich tumor microenvironment in cancer drug resistance, expanding our understanding of the roles of eATP in the Warburg effect and offering new anticancer drug resistance targets.


Cancer Research | 2017

Abstract 3194: A new mechanism of drug resistance in cancer: extracellular ATP-induced resistance through ATP internalization and upregulation of protein phosphorylation in Akt and ERK pathways

Xuan Wang; Yunsheng Li; Yanrong Qian; Yanyang Cao; Xiaozhuo Chen

The opportunistic uptake of extracellular molecules has been named as a key emerging hallmark of cancer metabolism[1]. Extracellular ATP (eATP) levels of various cancer types are 103 to 104 times higher than those in their corresponding normal tissues[2]. However, the biological significance of the high ATP concentrations is not clear. We recently reported that cancer cells uptake extracellular ATP via different endocytoses to enhance growth, survival, and drug resistance to tyrosine kinase inhibitors (TKIs)[3 ,4]. We hypothesized that eATP induces resistance to TKIs by endocytoses-mediated internalization of eATP, which competes with TKIs for the ATP-binding site of receptor TKs (RTKs), phosphorylating and activating RTKs and RTK-mediated signaling pathways. In contrast to previous reports that synthesized intracellular ATP (iATP) elevation contributed acquired drug resistance, here we report a novel intrinsic drug resistance in which eATP was internalized and substantially increased iATP levels and promoted cancer cell drug resistance to the TKI sunitinib in human NSCLC A549 cells. The iATP level elevation and drug resistance were mediated primarily by macropinocytosis. The resistance was reduced when macropinocytosis was suppressed by inhibitors or an siRNA knockdown of a key micropinocytosis protein PAK1. Intracellularly, the elevated iATP upregulated phosphorylation of PDGFR and proteins/enzymes in the PDGFR-mediated Akt-mTOR and Raf-ERK signaling pathways, resulting in reduced apoptosis triggered by sunitinib. Furthermore, both in vitro and in A549 tumors, eATP partially restored phosphorylation levels of PDGFR and PDGFR-mediated proteins/enzymes suppressed by sunitinib. The resistance cannot be accounted for by the overall purinergic receptor-mediated signaling, glycolysis, or mitochondrial OXPHOS. These results strongly suggest that the eATP-elevated intracellular ATP levels reversed the inhibition of TKIs by using the mechanism we hypothesized, linking for the first time the ATP-rich tumor microenvironment with cancer drug resistance. All these findings significantly expand our understanding of the roles of extracellular ATP in cancer, and offer new anti-drug resistance targets. 1. Pavlova N, Thompson C. Cell Metab. (2016) 23:27-47. 2. Falzoni et al., Interface Focus.(2013) 3: 20120101. 3. Qian Y, Wang X, et al., Cancer Lett. (2014)351: 242-5. 4. Qian Y, Wang X et al., Mol Cancer Res (2016) 14:1087-96 Citation Format: Xuan Wang, Yunsheng Li, Yanrong Qian, Yanyang Cao, Xiaozhuo Chen. A new mechanism of drug resistance in cancer: extracellular ATP-induced resistance through ATP internalization and upregulation of protein phosphorylation in Akt and ERK pathways [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3194. doi:10.1158/1538-7445.AM2017-3194


Cancer Research | 2016

Abstract 33: NSCLC cells internalize ATP in vitro and in vivo using multiple endocytotic pathways

Yanrong Qian; Xuan Wang; Yunsheng Li; Yanyang Cao; Xiaozhuo Chen

Upregulated glycolysis in cancer, the Warburg effect, has been studied for almost one century. However, the biological reasons for the upregulation are still debated and the effect is far from fully understood. One of the major controversies related to the effect is the role of ATP synthesis. Intratumoral ATP concentrations are found to be 10⁁3-10⁁4 times higher than those in normal tissues of the same cell origin. However, whether and how cancer cells use the abundant extracellular ATP was unknown until we recently reported that cancer cells internalize ATP by macropinocytosis (1). Extracellular ATP was found to increase intracellular ATP concentration and promote cell proliferation and drug resistance in cancer cells (1). Here we report that, using a nonhydrolyzable fluorescent ATP (NHF-ATP) as an ATP surrogate, high and low molecular weight dextrans as endocytosis tracers and fluorescence microscopy detection, cultured human NSCLC A549 and H1299 cells as well as xenografted A549 tumors were found to internalize ATP at concentrations within the reported intratumoral ATP concentrations. Endocytosis inhibitor studies show that in addition to macropinocytosis, clathrin- and caveolae-mediated endocytoses contribute to the ATP internalization, which led to a ∼30% increase in intracellular ATP level after 45 min of ATP incubation. This increase cannot be accounted for by purinergic receptor signaling or increased intracellular ATP synthesis rates. Internalization of NHF-ATP and dextran exhibits time-dependent profiles parallel to the extracellular ATP-induced intracellular ATP level elevation. All these demonstrate the observed ATP increase is a result of internalization of extracellular ATP and NHF-ATP is a powerful tool for studying ATP internalization. These findings significantly deepen our understanding of the Warburg effect by showing how cancer cells in tumors take up intratumoral ATP and perform previously unrecognized biological functions. It strongly suggests ATP-sharing among cancer and stromal cells and identifies multiple novel anticancer targets (2). 1. Qian Y, Wang X, et al., Cancer Lett. 351(2014): 242-5. 2. Chen X, Qian Y, Wu S. Free. Radic. Biol. Med. 79(2015): 253-263. Citation Format: Yanrong Qian, Xuan Wang, Yunsheng Li, Yanyang Cao, Xiaozhuo Chen. NSCLC cells internalize ATP in vitro and in vivo using multiple endocytotic pathways. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 33.


Cancer Research | 2016

Abstract 42: A new type of drug resistance in cancer: extracellular ATP-induced resistance through ATP internalization and ATP-drug competition

Xuan Wang; Yunsheng Li; Yanrong Qian; Yanyang Cao; Xiaozhuo Chen

Drug resistance is a major problem in cancer therapies and responsible for most relapse, one of the major causes of death in cancer. Several cancer drug resistance mechanisms are known, including genetic mutations in protooncogenes or tumor suppressor genes, cancer stem cells, and multi-drug resistance (MDR). Among these mechanisms, MDR is ATP-dependent. Intratumoral (extracellular) ATP levels of various cancer types are 10⁁3 to 10⁁4 times higher than those in their corresponding normal tissues but the biological significance of the high intratumoral ATP concentrations is unknown. We recently reported that extracellular ATP is internalized by cancer cells via macropinocytosis and enhances their growth and survival (1). Based on these results, we hypothesized that the internalized ATP also promotes drug resistance. Here we report that extracellular ATP, at concentrations in or below the range of reported intratumoral ATP levels, substantially increased intracellular ATP levels and promoted cancer cell drug resistance to tyrosine kinase inhibitor (TKI) sunitinib in human NSCLC A549 cells. The ATP increase and the drug resistance were mediated, at least in part, by macropinocytosis, clathrin- and caveolae-mediated endocytoses. The elevated intracellular ATP induced upregulated phosphorylation of PDGFR and proteins/enzymes in the PDGFR-mediated signaling pathways, such as Akt, mTOR, Raf and MEK, resulting in augmented cell growth and reduced apoptosis induced by sunitinib. The upregulated phosphorylation was not mediated by purinergic receptor signaling. Furthermore, in vitro, ex vivo, and in vivo, extracellular ATP partially restored phosphorylation levels of PDGRF and its downstream proteins/enzymes inhibited by sunitinib, a PDGFR inhibitor (TKI). These results strongly suggest that the extracellular ATP-increased intracellular ATP reversed the inhibition of TKIs by competing with the inhibitors for the ATP-binding site of PDGFR, enhancing the Akt-mTOR and Raf-MEK signaling pathways and promoting cancer cell survival. All these findings significantly expand our understanding of the roles of extracellular ATP in the Warburg effect (2) and drug resistance in cancer and indentify a new anti-drug resistance target. 1. Qian Y, Wang X, et al., Cancer Lett. 351(2014): 242-5. 2. Chen X, Qian Y, Wu S. Free. Radic. Biol. Med. 79(2015): 253-263. Citation Format: Xuan Wang, Yunsheng Li, Yanrong Qian, Yanyang Cao, Xiaozhuo Chen. A new type of drug resistance in cancer: extracellular ATP-induced resistance through ATP internalization and ATP-drug competition. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 42.


Cancer Research | 2015

Abstract 5462: Extracellular ATP promotes cancer cell drug resistance to tyrosine kinase inhibitors by competing for the ATP-binding site of tyrosine kinase

Xuan Wang; Yanrong Qian; Yunsheng Li; Xiaozhuo Chen

ATP plays important roles in cellular processes in cancer cells, which are a heterogeneous population in tumors regarding their genetic background and oxygen and energy supplies. The fact that cancer cells undergo rapid proliferation suggests that they must possess mechanisms, which may not be present in normal cells, to meet their high energy needs. Studies by others revealed that extracellular ATP levels of various cancer types are 10^3 to 10^4 times higher than those in their corresponding normal tissues (1). We recently reported that extracellular ATP can be taken up directly by cancer cells by macropinocytosis (2). Based on these results, we hypothesized that the internalized extracellular ATP not only promotes cancer cell growth and survival, but also drug resistance. In this study, human lung cancer A549 and breast cancer MCF7 cells were used as cell models to determine the effects of extracellular ATP on drug resistance to tyrosine kinase inhibitors (TKIs) in cancer cells. Cancer cell viability and intracellular ATP measurement results showed that extracellular ATP substantially increased intracellular ATP levels and promoted cancer cell drug resistance to TKIs. Viability of TKI treated cancer cells was significantly increased in the presence of extracellular ATP, compared to cancer cells with TKI treatment alone. Administration of macropinocytosis inhibitors significantly reduced the drug resistance induced by extracellular ATP, suggesting that extracellular ATP promoted drug resistance by contributing to the intracellular ATP pool. Protein and protein phosphorylation analyses results suggested that the increased intracellular ATP reversed the inhibition of TKIs by competing with the inhibitor for the ATP-binding site of platelet-derived growth factor receptor (PDGFR) and enhancing Akt and MAPK signaling pathways. These findings identify, for the first time, new biological functions of extracellular ATP in tumors and may lead to novel anti-cancer strategies combating drug resistance in cancers. 1. Pellegatti P, et al. PLoS ONE 2008;3:e2599. 2. Qian Y, et al. Cancer Lett 2014;351:242-51. Citation Format: Xuan Wang, Yanrong Qian, Yunsheng Li, Xiaozhuo Chen. Extracellular ATP promotes cancer cell drug resistance to tyrosine kinase inhibitors by competing for the ATP-binding site of tyrosine kinase. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5462. doi:10.1158/1538-7445.AM2015-5462


Cancer Research | 2014

Abstract 3364: Extracellular ATP-induced intracellular ATP concentration elevation mediated by macropinocytosis promotes growth, survival, and drug resistance of cancer cells

Yanrong Qian; Xuan Wang; Yi Liu; Yunsheng Li; Robert A. Colvin; Lingying Tong; Shiyong Wu; Xiaozhuo Chen

Cancer cells switch from highly efficient mitochondrial oxidative phosphorylation (OXPHOS) to low efficiency glycolysis for ATP synthesis even when oxygen is abundant, a phenomenon called the Warburg effect. Because of the higher rates of cell growth and proliferation, cancer cells need more ATP than normal cells of the same tissues. However, the switch is counterintuitive since cancer cells need more ATP but cancer cells prefer to use a low efficiency pathway to produce ATP. How cancer cells secure all their ATP needs is not well understood. On the other hand, extracellular ATP concentration in cancer (intratumoral ATP concentration) are found to be in the range of several hundred μM, ∼10^3-10^4 times higher than those found in normal tissues. Huge differences exist between cancer and normal cells regarding extracellular ATP. Based on these observations, we hypothesized that cancer cells take up ATP from extracellular space to supplement their energy needs. Here we report that in multiple cancer cell lines belonging to multiple cancer types, 0.5-3 mM ATP, within the intratumoral ATP concentration range, elevated intracellular ATP levels by more than 50%. Extracellular ATP also reduced stress and promoted survival of cancer cells that were under OXPHOS and glucose metabolism inhibitions. Furthermore, extracellular ATP increased the viability of cancer cells treated with sunitinib or pazopanib, anticancer drugs working as ATP analogs by competing with ATP for the ATP binding site of targeted tyrosine kinases (TKs). In contrast, extracellular ATP did not affect the activity of paclitaxel, a drug unrelated to ATP. Inhibitor studies revealed that the extracellular ATP-induced intracellular ATP increase shows no dependence on OXPHOS, glycolysis, master ATP regulator AMPK, transcription or translation. These results strongly suggest that not all the increased intracellular ATP is synthesized inside of cancer cells. Fluorescence microscopy study showed that human lung cancer A549 cells, which are KRas oncogene mutation-positive (KRas+), exhibit macropinotcytosis, a type of endocytosis that non-specifically take in extracellular nutritional molecules by “fluid drinking”. Inhibition of macropinocytosis resulted in decrease in dextran take-up and intracellular ATP levels, and alleviated the drug resistance to TKIs in dose-dependent manners. These strongly suggest that macropinocytosis is responsible, at least in part, for transporting extracellular ATP into cancer cells, which led to drug resistance. All these results demonstrate, for the first time, unrecognized roles of extracellular ATP in contributing to the intracellular ATP pool and increase drug resistance in cancer cells and suggest a novel ATP transport mechanism. These findings challenge our traditional view on ATP and shed new lights on ATP homeostasis and ATP-induced drug resistance in cancer cells. Citation Format: Yanrong Qian, Xuan Wang, Yi Liu, Yunsheng Li, Robert A. Colvin, Lingying Tong, Shiyong Wu, Xiaozhuo Chen. Extracellular ATP-induced intracellular ATP concentration elevation mediated by macropinocytosis promotes growth, survival, and drug resistance of cancer cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3364. doi:10.1158/1538-7445.AM2014-3364


Cancer Research | 2014

Abstract 5257: Cancer cells have not only sweet tooth but also large mouths: Evidence for uptake of ATP into KRas oncogene-expressing cancer cells by macropinocytosis

Yanrong Qian; Xuan Wang; Yi Liu; Yunsheng Li; Robert A. Colvin; Xiaozhuo Chen

Cancer cells are known to exhibit reprogrammed metabolism and prefer to use glycolygsis for their ATP synthesis even when oxygen is abundant, a phenomenon called the Warburg effect. However, cancer cells are also known for more rapid growth and proliferation, therefore requiring more ATP than normal cells of the same tissues. How cancer cells manage to use low efficiency glycolysis but still meet their higher ATP demands is enigmatic. In our previous study, we found that extracellular ATP induced large intracellular ATP increase and drug resistance to tyrosine kinase inhibitors (TKIs) that function as ATP analogs / competitors in cancer cells. The induced ATP increase was from neither OXPHOS nor glycolysis, and was not dependent on AMPK, the master ATP regulator. Furthermore, the increase did not involve new gene expression or new protein synthesis. Recently, it was reported that cells transformed with an oncogenic form of KRas gene (KRas+) exhibit a phenotype of macropinocytosis, a type of endocytosis. KRas-induced macropinocytosis nonspecifically takes up extracellular proteins. All these led us to hypothesize that the increased intracellular ATP induced by extracellular ATP is, at least in part, not produced intracellularly but is transported into cancer cells. To test this hypothesis, high molecular weight fluorescent dextran was use to determine if KRas+ human non-small cell lung cancer A549 cells exhibit macropinocytosis. Nonhydrolyzable fluorescent ATP analog was used to visualize if the ATP analog can be transported into A549 cells along with dextran. Fluorescence microscopy studies revealed that A549 cells exhibit the phenotype of macropinocytosis, which can be reduced by a macropinocytosis inhibitor EIPA. HMW dextran and the nonhydrolysable fluorescent ATP analog were found co-localized inside A549 cells, demonstrating that they were transported into A549 cells via macropinosomes. It was also found that the ATP increase cannot be explained by purinergic receptor signaling alone. In comparison, KRas- human breast cancer MCF7 cells do not take up HMW dextran, indicating that they do not exhibit macropinocytosis. All these provide strong and visual evidence that ATP can be transported into KRas+ A549 cancer cells by macropinocytosis. Other experiments using radioactive ATP or purinergic receptor inhibitors also strongly support this conclusion. This is the first time that ATP (ATP analog) is shown to be taken into cancer cells. This finding will have major impact on how we view cancer cells in terms of their energy exchange with other cancer or noncancer cells in the tumor and on the Warburg effect. It will also have profound implications on how to more effectively inhibiting KRas+ or KRas- cancer cells by targeting ATP transport and ATP metabolism. Citation Format: Yanrong Qian, Xuan Wang, Yi Liu, Yunsheng Li, Robert A. Colvin, Xiaozhuo Chen. Cancer cells have not only sweet tooth but also large mouths: Evidence for uptake of ATP into KRas oncogene-expressing cancer cells by macropinocytosis. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5257. doi:10.1158/1538-7445.AM2014-5257

Collaboration


Dive into the Yunsheng Li's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge