Network


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

Hotspot


Dive into the research topics where Jinyun Liu is active.

Publication


Featured researches published by Jinyun Liu.


Leukemia | 2008

A prognostic score for patients with lower risk myelodysplastic syndrome.

Guillermo Garcia-Manero; Jianquin Shan; S. Faderl; J. Cortes; Farhad Ravandi; G. Borthakur; William G. Wierda; Sherry Pierce; E. Estey; Jinyun Liu; Xuelin Huang; H. Kantarjian

Current prognostic models for myelodysplastic syndromes (MDS) do not allow the identification of patients with lower risk disease and poor prognosis that may benefit from early therapeutic intervention. We evaluated the characteristics of 856 patients with low or intermediate-1 disease by the International Prognostic Scoring System. Mean follow-up was 19.6 months (range 1–262). Of these patients, 87 (10%) transformed to acute myelogenous leukemia, and 429 (50%) had died. By multivariate analysis, characteristics associated with worse survival (P<0.01) were low platelets, anemia, older age, higher percent of marrow blasts and poor-risk cytogenetics. Although not included in the model, higher ferritin (P=0.007) and β2-microglobulin (P<0.001) levels were associated with worse prognosis. This allowed the development of a scoring system in which patients could be grouped in three categories: category 1 (n=182, 21%) with a median survival of 80.3 months (95% CI 68-NA); category 2 (n=408, 48%) with a median survival of 26.6 months (95% CI 22–32) and category 3 (n=265, 31%) with a median survival of 14.2 months (95% CI 13–18). In summary, this analysis indicates that it is possible to identify patients with lower risk MDS and poor prognosis who may benefit from early intervention.


Nature Cell Biology | 2012

Stromal control of cystine metabolism promotes cancer cell survival in chronic lymphocytic leukaemia

Wan Zhang; Dunyaporn Trachootham; Jinyun Liu; Gang Chen; Helene Pelicano; Celia Garcia-Prieto; Weiqin Lu; Jan A. Burger; Carlo M. Croce; William Plunkett; Michael J. Keating; Peng Huang

Tissue stromal cells interact with leukaemia cells and profoundly affect their viability and drug sensitivity. Here we show a biochemical mechanism by which bone marrow stromal cells modulate the redox status of chronic lymphocytic leukaemia (CLL) cells and promote cellular survival and drug resistance. Primary CLL cells from patients exhibit a limited ability to transport cystine for glutathione (GSH) synthesis owing to a low expression level of Xc-transporter. In contrast, bone marrow stromal cells effectively import cystine and convert it to cysteine, which is then released into the microenvironment for uptake by CLL cells to promote GSH synthesis. The elevated level of GSH enhances leukaemia cell survival and protects them from drug-induced cytotoxicity. Furthermore, disabling this protective mechanism significantly sensitizes CLL cells to drug treatment in the stromal environment. This stromal–leukaemia interaction is critical for CLL cell survival and represents a key biochemical pathway for effectively targeting leukaemia cells to overcome drug resistance in vivo.


Breast Cancer Research | 2014

Mitochondrial dysfunction in some triple-negative breast cancer cell lines: role of mTOR pathway and therapeutic potential.

Helene Pelicano; Wan Zhang; Jinyun Liu; Naima Hammoudi; Jiale Dai; Rui Hua Xu; Lajos Pusztai; Peng Huang

IntroductionTriple-negative breast cancer (TNBC) is a subtype of highly malignant breast cancer with poor prognosis. TNBC is not amenable to endocrine therapy and often exhibit resistance to current chemotherapeutic agents, therefore, further understanding of the biological properties of these cancer cells and development of effective therapeutic approaches are urgently needed.MethodsWe first investigated the metabolic alterations in TNBC cells in comparison with other subtypes of breast cancer cells using molecular and metabolic analyses. We further demonstrated that targeting these alterations using specific inhibitors and siRNA approach could render TNBC cells more sensitive to cell death compared to other breast cancer subtypes.ResultsWe found that TNBC cells compared to estrogen receptor (ER) positive cells possess special metabolic characteristics manifested by high glucose uptake, increased lactate production, and low mitochondrial respiration which is correlated with attenuation of mTOR pathway and decreased expression of p70S6K. Re-expression of p70S6K in TNBC cells reverses their glycolytic phenotype to an active oxidative phosphorylation (OXPHOS) state, while knockdown of p70S6K in ER positive cells leads to suppression of mitochondrial OXPHOS. Furthermore, lower OXPHOS activity in TNBC cells renders them highly dependent on glycolysis and the inhibition of glycolysis is highly effective in targeting TNBC cells despite their resistance to other anticancer agents.ConclusionsOur study shows that TNBC cells have profound metabolic alterations characterized by decreased mitochondrial respiration and increased glycolysis. Due to their impaired mitochondrial function, TNBC cells are highly sensitive to glycolytic inhibition, suggesting that such metabolic intervention may be an effective therapeutic strategy for this subtype of breast cancer cells.


Nature Medicine | 2017

Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth

Shira L. Cramer; Achinto Saha; Jinyun Liu; Surendar Tadi; Stefano Tiziani; Wupeng Yan; Kendra Triplett; Candice Lamb; Susan E. Alters; Scott W. Rowlinson; Yan Jessie Zhang; Michael J. Keating; Peng Huang; John DiGiovanni; George Georgiou; Everett M. Stone

Cancer cells experience higher oxidative stress from reactive oxygen species (ROS) than do non-malignant cells because of genetic alterations and abnormal growth; as a result, maintenance of the antioxidant glutathione (GSH) is essential for their survival and proliferation. Under conditions of elevated ROS, endogenous L-cysteine (L-Cys) production is insufficient for GSH synthesis. This necessitates uptake of L-Cys that is predominantly in its disulfide form, L-cystine (CSSC), via the xCT(−) transporter. We show that administration of an engineered and pharmacologically optimized human cyst(e)inase enzyme mediates sustained depletion of the extracellular L-Cys and CSSC pool in mice and non-human primates. Treatment with this enzyme selectively causes cell cycle arrest and death in cancer cells due to depletion of intracellular GSH and ensuing elevated ROS; yet this treatment results in no apparent toxicities in mice even after months of continuous treatment. Cyst(e)inase suppressed the growth of prostate carcinoma allografts, reduced tumor growth in both prostate and breast cancer xenografts and doubled the median survival time of TCL1-Tg:p53−/− mice, which develop disease resembling human chronic lymphocytic leukemia. It was observed that enzyme-mediated depletion of the serum L-Cys and CSSC pool suppresses the growth of multiple tumors, yet is very well tolerated for prolonged periods, suggesting that cyst(e)inase represents a safe and effective therapeutic modality for inactivating antioxidant cellular responses in a wide range of malignancies.Cancer cells experience higher oxidative stress from reactive oxygen species (ROS) than non-malignant cells due to genetic alterations and abnormal growth and as a result, maintenance of the anti-oxidant glutathione (GSH) is essential for their survival and proliferation1–3. Under elevated ROS conditions endogenous l-Cysteine (l-Cys) production is insufficient for GSH synthesis, necessitating l-Cys uptake, predominantly in its disulfide form l-Cystine (CSSC) via the xCT(−) transporter. Here we show that administration of an engineered, pharmacologically optimized, human Cyst(e)inase enzyme mediates sustained depletion of the extracellular l-Cys and CSSC pool in mice and non-human primates, selectively causes cell cycle arrest and death (PI and Annexin-V staining) in cancer cells due to depletion of intracellular GSH and ensuing elevated ROS, yet results in no apparent toxicities in mice even after months of continuous treatment. Cyst(e)inase suppressed the growth of prostate carcinoma allografts, reduced tumor growth in prostate and breast cancer xenografts and doubled the median survival time of TCL1-Tg:p53−/− mice that develop disease resembling human chronic lymphocytic leukemia. The observation that enzyme-mediated depletion of the serum l-Cys and CSSC pool suppresses the growth of multiple tumors, yet is very well tolerated for prolonged periods suggests that Cyst(e)inase represents a safe and effective therapeutic modality for inactivating anti-oxidant cellular responses in a wide range of malignancies4,5.


Stem Cell Research & Therapy | 2015

Metabolic activation of mitochondria in glioma stem cells promotes cancer development through a reactive oxygen species-mediated mechanism

Shuqiang Yuan; Yun-xin Lu; Jing Yang; Gang Chen; Sang Bae Kim; Li Feng; Marcia A. Ogasawara; Naima Hammoudi; Weiqin Lu; Hui Zhang; Jinyun Liu; Howard Colman; J. Lee; Xiao Nan Li; Ruihua Xu; Peng Huang; Feng Wang

IntroductionCancer stem cells (CSCs) possess characteristics associated with normal stem cells, specifically the abilities to renew themselves and to give rise to all cell types (differentiation). It is assumed that induction of differentiation in CSCs would reduce their ability to form tumors. What triggers CSC differentiation and the role of “differentiation” in tumorigenesis remain elusive.MethodsGlioma stem cell (GSC) lines and subcutaneous as well as orthotopic xenografts established from fresh surgical specimens of glioblastoma multiforme were used.ResultsExposure of GSCs to serum activates mitochondrial respiration and causes an increase in mitochondrial reactive oxygen species (ROS) as well as oxidative stress responses, leading to the appearance of differentiation morphology and a deceased expression of CSC markers. Chemical perturbation of the mitochondrial electron transport chain causes ROS increase and further downregulation of stem cell markers, while antioxidant N-acetyl-cysteine reduces ROS and suppresses the differentiation of GSCs. Surprisingly, the serum-induced differentiated GSCs exhibit greater ability to form tumor in both orthotopic and subcutaneous xenograft models, which can be suppressed by N-acetyl-cysteine. Mitochondrial ROS from the serum-stimulated cells triggered the activation of nuclear factor-kappa-B (NFκB) pathway, which is a potential mechanism for the promotion of tumorigenesis.ConclusionThis study suggests that ROS generated from active mitochondrial respiration in the presence of serum is critical in CSCs activation, which promotes tumor development in vivo.


Leukemia | 2014

Loss of p53 and altered miR15-a/16-1→MCL-1 pathway in CLL: Insights from TCL1-Tg:p53-/- mouse model and primary human leukemia cells

Jinyun Liu; Gang Chen; Li Feng; Wan Zhang; Helene Pelicano; Feng Wang; Marcia A. Ogasawara; Weiqin Lu; Hesham M. Amin; Carlo M. Croce; Michael J. Keating; Peng Huang

Chronic lymphocytic leukemia (CLL) patients with deletion of chromosome 17p, where the p53 gene is located, often develop more aggressive disease with poor clinical outcomes. To investigate the underlying mechanisms for the highly malignant phenotype of 17p- CLL and to facilitate in vivo evaluation of potential drugs against CLL with p53 deletion, we have generated a mouse model with TCL1-Tg:p53−/− genotype. These mice develop B-cell leukemia at an early age with an early appearance of CD5+/IgM+ B cells in the peritoneal cavity and spleen, and exhibit an aggressive path of disease development and drug resistance phenotype similar to human CLL with 17p deletion. The TCL1-Tg:p53−/− leukemia cells exhibit higher survival capacity and are more drug resistant than the leukemia cells from TCL1-Tg:p53wt mice. Analysis of microRNA expression reveals that p53 deletion resulted in a decrease of miR-15a and miR-16-1, leading to an elevated expression of Mcl-1. Primary leukemia cells from CLL patients with 17p deletion also show a decrease in miR-15a/miR-16-1 and an increase in Mcl-1. Our study suggests that the p53/miR15a/16-1/Mcl-1 axis may be an important pathway that regulates Mcl-1 expression and contributes to drug resistance and aggressive phenotype in CLL cells with loss of p53.


Oncotarget | 2016

Targeting p53-deficient chronic lymphocytic leukemia cells in vitro and in vivo by ROS-mediated mechanism

Jinyun Liu; Gang Chen; Helene Pelicano; Jianwei Liao; Jie Huang; Li Feng; Michael J. Keating; Peng Huang

Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in Western countries. Loss of p53 function in CLL cells due to chromosome 17p deletion or p53 mutations often leads to a more malignant disease phenotype and is associated with drug resistance and poor clinical outcome. Thus, development of novel therapeutic strategies to effectively target CLL cells with p53 deficiency is clinically important. Here we showed that p53-null CLL cells were highly sensitive to ROS-mediated cell killing due to their intrinsic ROS stress. We further demonstrated that a natural compound phenethyl isothiocyanate (PEITC) was able to effectively kill CLL cells with loss of p53, even under the protection of stromal cells. In p53-defficient CLL cells, PEITC induced a rapid depletion of glutathione and a severe accumulation of ROS, leading to massive leukemia cell death in the stromal microenvironment. The drug-induced cell death was associated with a significant decrease of in MCL-1 survival molecule. We further showed that ROS-mediated cell death was the key mechanism by which PEITC induced cytotoxicity, since such cell death could be prevented by addition of antioxidant NAC. Importantly, in vivo study showed that PEITC was able to induce substantial leukemia cell death in mice. Treatment of CLL mice harboring TCL1-Tg:p53−/− genotype with PEITC significantly prolonged the median survival time of the animals. Our study identifies a vulnerability of p53-null CLL cells with high sensitivity to ROS-generating agents, and suggests that PEITC may potentially be useful for clinical treatment of CLL with 17p deletion and p53 mutations.


Mitochondrion | 2016

Alterations of mitochondrial biogenesis in chronic lymphocytic leukemia cells with loss of p53

Marcia A. Ogasawara; Jinyun Liu; Helene Pelicano; Naima Hammoudi; Carlo M. Croce; Michael J. Keating; Peng Huang

Deletion of chromosome 17p with a loss of p53 is an unfavorable cytogenetic change in chronic lymphocytic leukemia (CLL) with poor clinical outcome. Since p53 affects mitochondrial function and integrity, we examined possible mitochondrial changes in CLL mice with TCL1-Tg/p53-/- and TCL1-Tg/p53+/+ genotypes and in primary leukemia cells from CLL patients with or without 17p-deletion. Although the expression of mitochondrial COX1, ND2, and ND6 decreased in p53-/-CLL cells, there was an increase in mitochondrial biogenesis as evidenced by higher mitochondrial mass and mtDNA copy number associated with an elevated expression of TFAM and PGC-1α. Surprisingly, the overall mitochondrial respiratory activity and maximum reserved capacity increased in p53-/- CLL cells. Our study suggests that leukemia cells lacking p53 seem able to maintain respiratory function by compensatory increase in mitochondrial biogenesis.


Cancer Research | 2016

Abstract 3073: Targeting chronic lymphocytic leukemia by interfering glutathione synthesis using a novel therapeutic enzyme cyst(e)inase (AEB3103)

Jinyun Liu; Li Feng; Everett M. Stone; Scott W. Rowlinson; Michael J. Keating; Peng Huang

Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in the Western countries. Despite recent advance in new therapeutic agents that have improved treatment outcomes, CLL remains incurable due in part to the inability to completely eradicate the leukemia cells in vivo. Previous studies showed that CLL cells have high intrinsic oxidative stress and are highly dependent on cellular antioxidant glutathione (GSH) to maintain redox balance and cell viability. One logical strategy to impact CLL cells would be to abrogate the glutathione protection of CLL cells in vivo. Recently we discovered that primary leukemia cells isolated from CLL patients were unable to effectively utilize cystine for GSH synthesis due to low expression of the cystine transporter Xc-, and that bone marrow stromal cells highly express Xc- and effectively take up cystine for conversion to cysteine (Zhang et al: Nature Cell Biology, 2012). These findings provide a biochemical basis to develop novel strategies to effectively target leukemia cells in the stromal microenvironment and improve in vivo therapeutic activity. In this study, we tested the depletion of extracellular cystine and cysteine using a novel therapeutic enzyme-cyst(e)inase (AEB3103), as a potential way to block GSH synthesis in CLL cells and abolish the stromal protection of the leukemia cells. Our study showed that AEB3103 was very effective in depleting GSH in CLL cells and caused massive CLL cell death even in the presence of stromal cells. Importantly, AEB3103 could also overcome drug resistance of CLL cells with p53 deficiency both in primary leukemia cells isolated from CLL patients with 17p deletion and mouse leukemic cells isolated from mouse model we recently reported (Liu et al: Leukemia, 2014). In addition, AEB3103 showed very low toxicity to normal cells. These promising in vitro data warrant further animal studies, which are currently on ongoing using the CLL mouse model with TCL1-Tg:p53-/- genotype. Our study suggests that AEB3103 and its combination with standard anti-CLL drugs may potentially be useful for clinical treatment of CLL, even for the more aggressive CLL subtypes with unfavorable cytogenetic alterations such as those with chromosome17p deletion and p53 mutations, and may improve in vivo therapeutic activity. Citation Format: Jinyun Liu, Li Feng, Everett M. Stone, Joseph Tyler, Scott W. Rowlinson, Michael J. Keating, Peng Huang. Targeting chronic lymphocytic leukemia by interfering glutathione synthesis using a novel therapeutic enzyme cyst(e)inase (AEB3103). [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 3073.


Cancer Research | 2014

Abstract 2697: A novel therapeutic strategy to effectively kill CLL cells in stromal microenvironment by targeting lipid metabolism

Panpan Liu; Jinyun Liu; Marcia A. Ogasawara; Helene Pelicano; Ruihua Xu; Michael J. Keating; Peng Huang

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA The tissue microenvironment promotes cancer cell survival and drug resistance, and thus presents a major challenge in cancer treatment. Recent study suggests that the microenvironmental protection of chronic lymphocytic leukemia (CLL) cells is particular important due to the unique metabolic properties of CLL cells. Emerging evidence suggests that altered fatty acid metabolism may be associated with the pathogenesis of CLL and could potentially provide a biochemical basis for targeting CLL. In the current study, we tested three classes of compounds that respectively inhibit the de novo fatty acids synthesis (FASN), block the transport of fatty acids from cytosol into mitochondria, or suppress fatty acid oxidation, for their ability to kill CLL cells in the presence and absence of bone marrow stromal cells. While all three inhibitors exhibited cytotoxic effect again CLL cells when the leukemia cells were culture alone, only the compound that inhibited the transport of fatty acids into mitochondria was highly effective in killing CLL in the presence of bone marrow stromal cells. Mechanistic study revealed that the cytotoxicity was not due to inhibition of ATP generation through mitochondrial β-oxidation of fatty acids, since there was no significant decrease in mitochondrial respiratory activity. We further showed that inhibition of FA transport into mitochondria suppressed biosynthesis of mitochondrial phospholipid, leading to a significant depletion of cardiolipin (CL), which is essential for maintaining mitochondrial membrane integrity. Depletion of CL by the FA transport inhibitor in CLL cells resulted in mitochondrial depolarization, release of cytochrome c, and massive apoptosis. Since this cell death process was through the intrinsic pathway and could not be rescued by the stromal environment, this might explain why inhibition of FA transport into the mitochondrial was highly effective in killing CLL cells in the presence of stromal cells. Importantly, we found that inhibition of FA transport had low toxicity in normal lymphocytes and stromal cells, suggesting that the normal cells might be less dependent on FA transport for cardiolipin biosynthesis. In summary, our study suggests that inhibition of FA transport into the mitochondria is a novel therapeutic strategy to effectively and selectively kill CLL cells in stromal microenvironment, and may potentially improve the clinical outcome of CLL treatment. Citation Format: Panpan Liu, Jinyun Liu, Marcia A. Ogasawara, Helene Pelicano, Ruihua Xu, Michael J. Keating, Peng Huang. A novel therapeutic strategy to effectively kill CLL cells in stromal microenvironment by targeting lipid metabolism. [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 2697. doi:10.1158/1538-7445.AM2014-2697

Collaboration


Dive into the Jinyun Liu's collaboration.

Top Co-Authors

Avatar

Peng Huang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael J. Keating

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Helene Pelicano

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gang Chen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Li Feng

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Marcia A. Ogasawara

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Wan Zhang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Everett M. Stone

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Naima Hammoudi

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge