Network


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

Hotspot


Dive into the research topics where Yuexi Shi is active.

Publication


Featured researches published by Yuexi Shi.


Stem Cells | 2010

Epithelial‐Mesenchymal Transition‐Derived Cells Exhibit Multilineage Differentiation Potential Similar to Mesenchymal Stem Cells

Venkata Lokesh Battula; Kurt W. Evans; Brett G. Hollier; Yuexi Shi; Frank C. Marini; Ayyakkannu Ayyanan; Rui Yu Wang; Cathrin Brisken; Rudy Guerra; Michael Andreeff; Sendurai A. Mani

The epithelial‐to‐mesenchymal transition (EMT) is an embryonic process that becomes latent in most normal adult tissues. Recently, we have shown that induction of EMT endows breast epithelial cells with stem cell traits. In this report, we have further characterized the EMT‐derived cells and shown that these cells are similar to mesenchymal stem cells (MSCs) with the capacity to differentiate into multiple tissue lineages. For this purpose, we induced EMT by ectopic expression of Twist, Snail, or transforming growth factor‐β in immortalized human mammary epithelial cells. We found that the EMT‐derived cells and MSCs share many properties including the antigenic profile typical of MSCs, that is, CD44+, CD24−, and CD45−. Conversely, MSCs express EMT‐associated genes, such as Twist, Snail, and mesenchyme forkhead 1 (FOXC2). Interestingly, CD140b (platelet‐derived growth factor receptor‐β), a marker for naive MSCs, is exclusively expressed in EMT‐derived cells and not in their epithelial counterparts. Moreover, functional analyses revealed that EMT‐derived cells but not the control cells can differentiate into alizarin red S‐positive mature osteoblasts, oil red O‐positive adipocytes and alcian blue‐positive chondrocytes similar to MSCs. We also observed that EMT‐derived cells but not the control cells invade and migrate towards MDA‐MB‐231 breast cancer cells similar to MSCs. In vivo wound homing assays in nude mice revealed that the EMT‐derived cells home to wound sites similar to MSCs. In conclusion, we have demonstrated that the EMT‐derived cells are similar to MSCs in gene expression, multilineage differentiation, and ability to migrate towards tumor cells and wound sites. STEM CELLS 2010;28:1435–1445


Cytotherapy | 2010

Mesenchymal stromal cells alone or expressing interferon-β suppress pancreatic tumors in vivo, an effect countered by anti-inflammatory treatment

Shannon Kidd; Lisa Caldwell; Martin Dietrich; Ismael Samudio; Erika L. Spaeth; Keri Watson; Yuexi Shi; James L. Abbruzzese; Marina Konopleva; Michael Andreeff; Frank C. Marini

BACKGROUND AIMS Because of the inflammatory nature and extensive stromal compartment in pancreatic tumors, we investigated the role of mesenchymal stromal cells (MSC) to engraft selectively in pancreatic carcinomas and serve as anti-tumor drug delivery vehicles to control pancreatic cancer progression. METHODS Human pancreatic carcinoma cells, PANC-1, expressing renilla luciferase were orthotopically implanted into SCID mice and allowed to develop for 10 days. Firefly luciferase-transduced MSC or MSC expressing interferon (IFN)-beta were then injected intraperitoneally weekly for 3 weeks. Mice were monitored by bioluminescent imaging for expression of renilla (PANC-1) and firefly (MSC) luciferase. RESULTS MSC selectively homed to sites of primary and metastatic pancreatic tumors and inhibited tumor growth (P=0.032). The production of IFN-beta within the tumor site by MSC-IFN-beta further suppressed tumor growth (P=0.0000083). Prior studies indicated that MSC home to sites of inflammation; therefore, we sought to alter the tumor microenvironment through treatment with a potent anti-inflammatory agent. After treatment, inflammation-associated mediators were effectively down-regulated, including NFkappaB, vascular endothelial growth factor (VEGF) and interleukin (IL)-6 as well as chemokines involved in MSC migration (CCL3 and CCL25). Treatment with the anti-inflammatory agent CDDO-Me before and after MSC-IFN-beta injections resulted in reduction of MSC in the tumors and reversed the positive effect of tumor inhibition by MSC-IFN-beta alone (P=0.041). CONCLUSIONS These results suggest that MSC exhibit innate anti-tumor effects against PANC-1 cells and can serve as delivery vehicles for IFN-beta for the treatment of pancreatic cancer. However, these beneficial effects may be lost in therapies combining MSC with anti-inflammatory agents.


Journal of Clinical Investigation | 2012

Ganglioside GD2 identifies breast cancer stem cells and promotes tumorigenesis

Venkata Lokesh Battula; Yuexi Shi; Kurt W. Evans; Rui Yu Wang; Erika L. Spaeth; Rodrigo Jacamo; Rudy Guerra; Aysegul A. Sahin; Frank C. Marini; Gabriel N. Hortobagyi; Sendurai A. Mani; Michael Andreeff

Cancer stem cells (CSCs) are a small subpopulation of cancer cells that have increased resistance to conventional therapies and are capable of establishing metastasis. However, only a few biomarkers of CSCs have been identified. Here, we report that ganglioside GD2 (a glycosphingolipid) identifies a small fraction of cells in human breast cancer cell lines and patient samples that are capable of forming mammospheres and initiating tumors with as few as 10 GD2+ cells. In addition, the majority of GD2+ cells are also CD44hiCD24lo, the previously established CSC-associated cell surface phenotype. Gene expression analysis revealed that GD3 synthase (GD3S) is highly expressed in GD2+ as well as in CD44hiCD24lo cells and that interference with GD3S expression, either by shRNA or using a pharmacological inhibitor, reduced the CSC population and CSC-associated properties. GD3S knockdown completely abrogated tumor formation in vivo. Also, induction of epithelial-mesenchymal transition (EMT) in transformed human mammary epithelial cells (HMLER cells) dramatically increased GD2 as well as GD3S expression in these cells, suggesting a role of EMT in the origin of GD2+ breast CSCs. In summary, we identified GD2 as a new CSC-specific cell surface marker and GD3S as a potential therapeutic target for CSCs, with the possibility of improving survival and cure rates in patients with breast cancer.


PLOS ONE | 2011

Pronounced hypoxia in models of murine and human leukemia: high efficacy of hypoxia-activated prodrug PR-104.

Juliana Benito; Yuexi Shi; Barbara Szymanska; Hernan Carol; Ingrid Boehm; Hongbo Lu; Sergej Konoplev; Wendy Fang; Patrick A. Zweidler-McKay; Dario Campana; Gautam Borthakur; Carlos E. Bueso-Ramos; Elizabeth J. Shpall; Deborah A. Thomas; Craig T. Jordan; Hagop M. Kantarjian; William R. Wilson; Richard B. Lock; Michael Andreeff; Marina Konopleva

Recent studies indicate that interactions between leukemia cells and the bone marrow (BM) microenvironment promote leukemia cell survival and confer resistance to anti-leukemic drugs. There is evidence that BM microenvironment contains hypoxic areas that confer survival advantage to hematopoietic cells. In the present study we investigated whether hypoxia in leukemic BM contributes to the protective role of the BM microenvironment. We observed a marked expansion of hypoxic BM areas in immunodeficient mice engrafted with acute lymphoblastic leukemia (ALL) cells. Consistent with this finding, we found that hypoxia promotes chemoresistance in various ALL derived cell lines. These findings suggest to employ hypoxia-activated prodrugs to eliminate leukemia cells within hypoxic niches. Using several xenograft models, we demonstrated that administration of the hypoxia-activated dinitrobenzamide mustard, PR-104 prolonged survival and decreased leukemia burden of immune-deficient mice injected with primary acute lymphoblastic leukemia cells. Together, these findings strongly suggest that targeting hypoxia in leukemic BM is feasible and may significantly improve leukemia therapy.


Journal of the National Cancer Institute | 2014

Synergistic Targeting of AML Stem/Progenitor Cells With IAP Antagonist Birinapant and Demethylating Agents

Bing Z. Carter; Po Yee Mak; Duncan H. Mak; Yuexi Shi; Yihua Qiu; James M Bogenberger; Hong Mu; Raoul Tibes; Hui Yao; Kevin R. Coombes; Rodrigo Jacamo; Teresa McQueen; Steven M. Kornblau; Michael Andreeff

BACKGROUND Acute myeloid leukemia (AML) therapy has limited long-term efficacy because patients frequently develop disease relapse because of the inability of standard chemotherapeutic agents to target AML stem/progenitor cells. Here, we identify deregulated apoptotic components in AML stem/progenitor cells and investigate the individual and combinatorial effects of the novel inhibitor of apoptosis (IAP) protein antagonist and second mitochondrial-derived activator of caspases (SMAC) mimetic birinapant and demethylating epigenetic modulators. METHODS Protein expression was measured by reversed-phase protein array in AML patient (n = 511) and normal (n = 21) samples and by western blot in drug-treated cells. The antileukemic activity of birinapant and demethylating agents was assessed in vitro and in an in vivo AML mouse xenograft model (n = 10 mice per group). All statistical tests were two-sided. RESULTS Compared with bulk AML cells, CD34(+)38(-) AML stem/progenitors expressed increased cIAP1 and caspase-8 levels and decreased SMAC levels (one-way analysis of variance followed by Tukeys multiple comparison test, P < .001). Birinapant induced death receptor-/caspase-8-mediated apoptosis in AML cells, including in AML stem/progenitor cells, but not in normal CD34(+) cells. Demethylating agents modulated extrinsic apoptosis pathway components and, when combined with birinapant, were highly synergistic in vitro (combination index < 1), and also more effective in vivo (P < .001, by Student t test, for the median survival of birinapant plus 5-azacytadine vs birinapant alone or vs controls). CONCLUSIONS cIAP1, SMAC, and caspase-8 appear to play a role in AML stem cell survival, and synergistic targeting of these cells with birinapant and demethylating agents shows potential utility in leukemia therapy.


PLOS ONE | 2013

The CXCR4 Antagonist AMD3465 Regulates Oncogenic Signaling and Invasiveness In Vitro and Prevents Breast Cancer Growth and Metastasis In Vivo

Xiaoyang Ling; Erika L. Spaeth; Ye Chen; Yuexi Shi; Weiguo Zhang; Wendy D. Schober; Numsen Hail; Marina Konopleva; Michael Andreeff

CXCR4, the receptor for stromal-derived factor-1, is reportedly involved in breast carcinogenesis. However, the mechanisms through which CXCR4 contributes to breast cancer cell growth and metastases are poorly understood. In this study, we examined the putative in vitro and in vivo anti-cancer effects of the specific CXCR4 inhibitor AMD3465. Here, we report that AMD3465 triggers a reduction in breast cancer cell invasiveness in vitro, and promotes marked changes in oncogenic signaling proteins including a reduction in STAT3, JAK2, AKT, and CXCR4 phosphorylation and the reduced expression of GSK3 and cMYC. Using three breast cancer cell lines as murine syngeneic immunocompetent breast cancer models, we found that AMD3465 inhibited breast tumor formation and reduced tumor cell metastases to the lung and liver. Furthermore, treatment with AMD3465 significantly reduced the infiltration of myeloid CD11b positive cells at the aforementioned metastatic sites as well as the spleen implying this agent could regulate the formation of the tumor microenvironment and conceivably the premetastatic niche. In conclusion, our studies suggest that AMD3465 inhibits breast cancer growth and metastases by acting on tumor cells as well as immune cells that constitute the tumor microenvironment. This process appears to be regulated, at least in part, through the modulation of oncogenic signaling that includes the STAT3 pathway. Thus, CXCR4 could be a novel target for breast cancer therapy.


Cell Cycle | 2006

Regulation and targeting of Eg5, a mitotic motor protein in blast crisis CML: Overcoming imatinib resistance

Bing Z. Carter; Duncan H. Mak; Yuexi Shi; Wendy D. Schober; Rui Yu Wang; Marina Konopleva; Erich Koller; Nicholas M. Dean; Michael Andreeff

Patients with blast crisis (BC) CML frequently become resistant to Imatinib, a Bcr-Abltyrosine kinase-targeting agent. Eg5, a microtubule-associated motor protein has beendescribed to be highly expressed in BC CML by microarray analysis (Nowicki et al,Oncogene 22:3952-3963, 2003). We investigated the regulation of Eg5 by Bcr-Abltyrosine kinase and its potential as a therapeutic target in BC CML. Eg5 was highlyexpressed in all Philadelphia chromosome positive (Ph+) cell lines and BC CML patientsamples. Inhibition of Bcr-Abl by Imatinib downregulated Eg5 expression in ImatinibsensitiveKBM5 and HL-60p185 cells, but not in Imatinib-resistant KBM5-STI571,harboring a T315I mutation, and Bcr-Abl-negative HL-60 cells. Blocking Eg5 expressionwith antisense oligonucleotide (Eg5-ASO) or inhibiting its activity with the smallmoleculeEg5 inhibitor, S-trityl-L-cysteine induced G2/M cell cycle block and subsequentcell death in both Imatinib-sensitive and -resistant cells. Further, Eg5-ASO treatment ofSCID mice harboring KBM5 cell xenografts significantly prolonged the median survivalof the animals (p=0.03). Our findings suggest that Eg5 is downstream of and regulated byBcr-Abl tyrosine kinase in Philadelphia chromosome positive cells. Inhibition of Eg5expression or its activity blocks cell cycle progression and induces cell death independentof the cellular response to Imatinib. Therefore, Eg5 could be a potential therapeutic targetfor the treatment of BC CML, in particular Imatinib-resistant BC CML.


Cancer Biology & Therapy | 2007

PPARgamma-Active triterpenoid CDDO enhances ATRA-induced differentiation in APL

Yoko Tabe; Marina Konopleva; Yutaka Kondo; Rooha Contractor; Twee Tsao; Sergej Konoplev; Yuexi Shi; Xiaoyang Ling; Julie C. Watt; Yuko Tsutsumi-Ishii; Akimichi Ohsaka; Isao Nagaoka; Jean-Pierre Issa; Scott C. Kogan; Michael Andreeff

Acute promyelocytic leukemia (APL) is associated with oncogenic PML-RARα that acts as a dominant negative transcriptional repressor of retinoic acid (RA) receptor target genes by recruiting histone deacetylase (HDAC). The peroxisome proliferator-activated receptor-γ (PPARγ) is a member of the nuclear receptor family that forms heterodimers with retinoid X receptor (RXR). In addition to RAR targets, PML-RARα silence a wide range of nuclear receptor target genes including PPARγ targets. All-trans-retinoic acid (ATRA), a ligand for the RA receptor (RAR), restores normal retinoid signaling and induces terminal differentiation of APL cells; however, APL cells can develop resistance to ATRA. Using ATRA sensitive NB4 and ATRA-resistant derivative NB4MR2 cell lines, we demonstrate that PPARγ ligand 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) enhances pro-apoptotic and differentiating effects of ATRA in ATRA-sensitive NB4 cells and partially reverses ATRA resistance in NB4MR2 cells. The CDDO/ATRA combination synergistically induces RARβ2 expression both in ATRA-sensitive and -resistant APL cells. RARβ2 mRNA induction by CDDO/ATRA was mediated in part by enhanced H3-Lys9 acetylation in the RARβ2 promoter which in turn increased the affinity of RARβ for βRARE. PPARγ specific inhibitor T007 and silencing of PPARγ by siRNA diminished CDDO-induced maturation and RARβ2 mRNA along with PPARγ induction indicating that PPARγ activation is at least partially responsible for the RARβ2 transcription and maturation induction. In an in vivo mouse model of APL, CDDO derivative CDDO-methyl ester markedly enhanced ATRA-induced maturation and extended the survival of mice. In summary, these results provide rationale for the combined targeting of RAR and PPARγ nuclear receptors in the therapy of APL.


Annals of Hematology | 2012

Concomitant inhibition of DNA methyltransferase and BCL-2 protein function synergistically induce mitochondrial apoptosis in acute myelogenous leukemia cells

Twee Tsao; Yuexi Shi; Steven M. Kornblau; Hongbo Lu; Sergej Konoplev; Ansu Antony; Vivian Ruvolo; Yi Hua Qiu; Ninaxiang Zhang; Kevin R. Coombes; Michael Andreeff; Kensuke Kojima; Marina Konopleva

DNA methylation and BLC-2 are potential therapeutic targets in acute myeloid leukemia (AML). We investigated pharmacologic interaction between the DNA methyltransferase inhibitor 5-azacytidine (5-AZA) and the BCL-2 inhibitor ABT-737. Increased BCL-2 expression determined by reverse phase protein analysis was associated with poor survival in AML patients with unfavorable cytogenetics (n = 195). We found that 5-AZA, which itself has modest apoptotic activity, acts synergistically with ABT-737 to induce apoptosis. The 5-AZA/ABT-737 combination enhanced mitochondrial outer membrane permeabilization, as evidenced by effective conformational activation of BAX and ∆ψm loss. Although absence of p53 limited apoptotic activities of 5-AZA and ABT-737 as single agents, the combination synergistically induced apoptosis independent of p53 expression. 5-AZA down-regulated MCL-1, known to mediate resistance to ABT-737, in a p53-independent manner. The 5-AZA/ABT-737 combination synergistically induced apoptosis in AML cells in seven of eight patients. 5-AZA significantly reduced MCL-1 levels in two of three samples examined. Our data provide a molecular rationale for this combination strategy in AML therapy.


Annals of Hematology | 2014

Targeting connective tissue growth factor (CTGF) in acute lymphoblastic leukemia preclinical models: Anti-CTGF monoclonal antibody attenuates leukemia growth

Hongbo Lu; Kensuke Kojima; Venkata Lokesh Battula; Borys Korchin; Yuexi Shi; Ye Chen; Suzanne Spong; Deborah A. Thomas; Hagop M. Kantarjian; Richard B. Lock; Michael Andreeff; Marina Konopleva

Connective tissue growth factor (CTGF/CCN2) is involved in extracellular matrix production, tumor cell proliferation, adhesion, migration, and metastasis. Recent studies have shown that CTGF expression is elevated in precursor B-acute lymphoblastic leukemia (ALL) and that increased expression of CTGF is associated with inferior outcome in B-ALL. In this study, we characterized the functional role and downstream signaling pathways of CTGF in ALL cells. First, we utilized lentiviral shRNA to knockdown CTGF in RS4;11 and REH ALL cells expressing high levels of CTGF mRNA. Silencing of CTGF resulted in significant suppression of leukemia cell growth compared to control vector, which was associated with AKT/mTOR inactivation and increased levels of cyclin-dependent kinase inhibitor p27. CTGF knockdown sensitized ALL cells to vincristine and methotrexate. Treatment with an anti-CTGF monoclonal antibody, FG-3019, significantly prolonged survival of mice injected with primary xenograft B-ALL cells when co-treated with conventional chemotherapy (vincristine, L-asparaginase and dexamethasone). Data suggest that CTGF represents a targetable molecular aberration in B-ALL, and blocking CTGF signaling in conjunction with administration of chemotherapy may represent a novel therapeutic approach for ALL patients.

Collaboration


Dive into the Yuexi Shi's collaboration.

Top Co-Authors

Avatar

Michael Andreeff

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Marina Konopleva

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Teresa McQueen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sergej Konoplev

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hongbo Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rodrigo Jacamo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven M. Kornblau

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Bing Z. Carter

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Juliana Benito

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge