Browsing by Author "Ding, Yi"
Results Per Page
Sort Options
Item Open Access Distinct Receptor Tyrosine Kinase Subsets Mediate Anti-HER2 Drug Resistance in Breast Cancer.(J Biol Chem, 2017-01-13) Alexander, Peter B; Chen, Rui; Gong, Chang; Yuan, Lifeng; Jasper, Jeff S; Ding, Yi; Markowitz, Geoffrey J; Yang, Pengyuan; Xu, Xin; McDonnell, Donald P; Song, Erwei; Wang, Xiao-FanTargeted inhibitors of the human epidermal growth factor receptor 2 (HER2), such as trastuzumab and lapatinib, are among the first examples of molecularly targeted cancer therapy and have proven largely effective for the treatment of HER2-positive breast cancers. However, approximately half of those patients either do not respond to these therapies or develop secondary resistance. Although a few signaling pathways have been implicated, a comprehensive understanding of mechanisms underlying HER2 inhibitor drug resistance is still lacking. To address this critical question, we undertook a concerted approach using patient expression data sets, HER2-positive cell lines, and tumor samples biopsied both before and after trastuzumab treatment. Together, these methods revealed that high expression and activation of a specific subset of receptor tyrosine kinases (RTKs) was strongly associated with poor clinical prognosis and the development of resistance. Mechanistically, these RTKs are capable of maintaining downstream signal transduction to promote tumor growth via the suppression of cellular senescence. Consequently, these findings provide the rationale for the design of therapeutic strategies for overcoming drug resistance in breast cancer via combinational inhibition of the limited number of targets from this specific subset of RTKs.Item Open Access Metabolic vulnerability in HER2-positive Breast Cancer(2018) Ding, YiThe human epidermal growth factor receptor 2, or HER2, is overexpressed in 20-30% breast cancer patients and is associated with aggressive disease. Therapies targeting HER2, including monoclonal antibodies (trastuzumab and pertuzumab), a small molecule kinase inhibitor (lapatinib) and an antibody-drug conjugate (trastuzumab emtansine), have significantly prolonged the overall survival of HER2-positive breast cancer patients. However, almost all patients develop resistance either from the beginning of therapy or with prolonged treatment in two years.
Previous studies to unveil the resistance mechanisms were mainly focused on acquired resistance, culturing cells with HER2 inhibitors and making comparisons to their parental cells. In order to study the mechanism mediating intrinsic resistance, we conducted a loss-of-function genetic screen using a HER2-amplified cell line that is intrinsically resistant to HER2 inhibitors with the purpose to identify synthetic lethal targets. TALDO1, a gene encoding a metabolic enzyme in the non-oxidative pentose phosphate pathway was identified from the screen. Metabolic profiling with isotope-labeled glucose was used to understand the mechanism. The profiling results indicated that TALDO1 was necessary for cellular NADPH generation to combat increased cellular ROS and support synthesis of lipids as a result of HER2 inhibition.
Importantly, the higher expression of TALDO1 is associated with poor response to HER2-targeted therapy in a small cohort of HER2-positive breast cancer patients, suggesting it could potentially serve as a biomarker to predict patient response.
Together our study explained a novel mechanism mediating intrinsic resistance to HER2 inhibition with significant clinical value. Combined inhibition of HER2 signaling and the pentose phosphate pathway may result in a better clinical outcome.
Item Open Access Synthetic lethality between HER2 and transaldolase in intrinsically resistant HER2-positive breast cancers.(Nature communications, 2018-10) Ding, Yi; Gong, Chang; Huang, De; Chen, Rui; Sui, Pinpin; Lin, Kevin H; Liang, Gehao; Yuan, Lifeng; Xiang, Handan; Chen, Junying; Yin, Tao; Alexander, Peter B; Wang, Qian-Fei; Song, Er-Wei; Li, Qi-Jing; Wood, Kris C; Wang, Xiao-FanIntrinsic resistance to anti-HER2 therapy in breast cancer remains an obstacle in the clinic, limiting its efficacy. However, the biological basis for intrinsic resistance is poorly understood. Here we performed a CRISPR/Cas9-mediated loss-of-function genetic profiling and identified TALDO1, which encodes the rate-limiting transaldolase (TA) enzyme in the non-oxidative pentose phosphate pathway, as essential for cellular survival following pharmacological HER2 blockade. Suppression of TA increases cell susceptibility to HER2 inhibition in two intrinsically resistant breast cancer cell lines with HER2 amplification. Mechanistically, TA depletion combined with HER2 inhibition significantly reduces cellular NADPH levels, resulting in excessive ROS production and deficient lipid and nucleotide synthesis. Importantly, higher TA expression correlates with poor response to HER2 inhibition in a breast cancer patient cohort. Together, these results pinpoint TA as a novel metabolic enzyme possessing synthetic lethality with HER2 inhibition that can potentially be exploited as a biomarker or target for combination therapy.Item Open Access UHRF1 is required for basal stem cell proliferation in response to airway injury.(Cell Discov, 2017) Xiang, Handan; Yuan, Lifeng; Gao, Xia; Alexander, Peter B; Lopez, Omar; Lau, Calvin; Ding, Yi; Chong, Mengyang; Sun, Tao; Chen, Rui; Liu, Si-Qi; Wu, Haiyang; Wan, Ying; Randell, Scott H; Li, Qi-Jing; Wang, Xiao-FanCellular senescence is a cell fate characterized by an irreversible cell cycle arrest, but the molecular mechanism underlying this senescence hallmark remains poorly understood. Through an unbiased search for novel senescence regulators in airway basal cells, we discovered that the epigenetic regulator ubiquitin-like with PHD and ring finger domain-containing protein 1 (UHRF1) is critical for regulating cell cycle progression. Upon injury, basal cells in the mouse airway rapidly induce the expression of UHRF1 in order to stimulate stem cell proliferation and tissue repair. Targeted depletion of Uhrf1 specifically in airway basal cells causes a profound defect in cell cycle progression. Consistently, cultured primary human basal cells lacking UHRF1 do not exhibit cell death or differentiation phenotypes but undergo a spontaneous program of senescence. Mechanistically, UHRF1 loss induces G1 cell cycle arrest by abrogating DNA replication factory formation as evidenced by loss of proliferating cell nuclear antigen (PCNA) puncta and an inability to enter the first cell cycle. This proliferation defect is partially mediated by the p15 pathway. Overall, our study provides the first evidence of an indispensable role of UHRF1 in somatic stem cells proliferation during the process of airway regeneration.