2018 Harvard Chinese Life Sciences YONGJIN Distinguished Research Award Awardees

Apr - 30
2018

2018 Harvard Chinese Life Sciences YONGJIN Distinguished Research Award Awardees

Harvard Chinese Life Sciences (HCLS) YONGJIN Distinguished Research Award is sponsored by Yongjin Group, which is to reward Chinese scientists whose research and discoveries were published or accepted for publication in 2017 & 2018 and well recognized by the scientific community. After tons of applications and some seriously stiff competition, we’re super excited to announce that the following twelve researchers are awarded 2018 Harvard Chinese Life Sciences YONGJIN Distinguished Research Award! 

They will be awarded at the Harvard Chinese Life Science Annual Research Symposium on May 5, 2018, the symposium will take place at the Folkman Auditorium, Enders Building, Boston Children’s Hospital, 320 Longwood Ave, Boston, MA 02115.

Chongyi Chen, Ph.D. 

Department of Chemistry and Chemical Biology, Harvard University

Dr. Chen is a postdoctoral fellow from Prof. Xiaoliang Sunney Xie’s lab at Harvard University, studying single-cell genomics. He developed a single-cell whole-genome amplification (WGA) method, Linear Amplification via Transposon Insertion (LIANTI). The new method supersedes previous single-cell WGA methods in various key aspects, enabling the detection of single-cell copy-number variations genome-wide at kilobase resolution. The LIANTI method was recently published in Science (2017), with many ongoing efforts at the moment in relevant technology development, as well as scientific and clinical applications.

Publication (2017~2018):

  • Chongyi Chen*, Dong Xing*, Longzhi Tan*, Heng Li*, Guangyu Zhou, Lei Huang, X. Sunney Xie. “Single-cell Whole-genome Analyses by Linear Amplification via Transposon Insertion (LIANTI)”, Science 356, 189-194 (2017) (*co-first author)

Kangmin He, Ph.D.

Boston Children’s Hospital, Harvard Medical School

Dr. He received his Ph.D. from Peking University in 2013 and then started his postdoc training in Dr. Tom Kirchhausen’s Lab at Boston Children’s Hospital and Harvard Medical School. Currently he is a research associate at Boston Children’s Hospital and instructor at Harvard Medical School. His research is characterized by use of advanced optical-imaging microscopies to examine the cell membrane trafficking processes. Especially his current research has focused on studying the molecular mechanisms that underlie the assembly and dissemble of clathrin-coated molecular machineries using quantitative live-cell imaging techniques. 

Publication (2017~2018):

  • Kangmin He+, Robert Marsland III, Srigokul Upadhyayula, Eli Song, Song Dang, Ben Capraro, Weiming Wang, Wesley Skillern, Raphael Gaudin, Minghe Ma, Tom Kirchhausen+. Dynamics of phosphoinositide conversion in clathrin-mediated endocytic traffic. Nature (2017) 552:410-414. (+co-corresponding author)
  • Nan Li, Rong Zhao, Yahong Sun, Zi Ye, Kangmin He+, Xiaohong Fang+. Single-molecule imaging and tracking of molecular dynamics in living cells. National Science Review (2017) 4:739–760. (+co-corresponding author)

Contact Information: Kangmin.He (at) childrens.harvard.edu

Julia Joung, Ph.D. Candidate

Broad Institute of MIT and Harvard
Department of Biological Engineering, MIT

Julia Joung is a graduate student in the lab of Feng Zhang at the Broad Institute of MIT and Harvard. She received her B.S. from Stanford University in bioengineering, where she worked in the neurobiology lab of Ben Barres on astrocyte-mediated synaptic pruning. At Stanford, she was selected for the departmental commencement speaker and for the Terman Engineering Scholastic Award. After graduation, as a research scientist at Counsyl, she helped develop a large-scale, cost-effective sequencing protocol for genetic diagnostics. In the lab of Feng Zhang, she develops CRISPR-Cas9 tools for studying long noncoding RNAs and neurological diseases.

Publication (2017~2018): 

  • Joung, J., Engreitz, J.M., Konermann, S., Abudayyeh, O.O., Verdine, V.K., Aguet, F., Gootenberg, J.S., Sanjana, N.E., Wright, J.B., Fulco, C.P., Tseng, Y.Y., Yoon, C.H., Boehm, J.S., Lander, E.S., and Zhang, F. “Genome-scale activation screen identifies a lncRNA locus regulating a gene neighborhood,” Nature, 2017.
  • Joung, J., Konermann, S., Gootenberg, J.S., Abudayyeh, O.O., Platt, R.J., Brigham, M.D., Sanjana, N.E., and Zhang, F. “Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening,” Nature Protocols, 2017.

Ying Li, Ph.D.

Department of Molecular and Cellular Biology, Harvard University

Dr. Li currently a fifth-year postdoctoral fellow in the laboratory of Prof. Catherine Dulac in the Department of Molecular and Cellular Biology, Harvard University. In 2013, she graduated from the Institute of Neuroscience, Chinese Academy of Science, Shanghai where she earned a Ph.D degree in neuroscience with Dr. Jiulin Du. Her primary interest is to understand the neural basis of social information processing in instinctive circuitry. One of her major contributions is to reveal the neural principles of social representation in the medial amygdala (MeA), a key hub of the sensory-motor transformation from chemosensory cues to instinctive behaviors in mice (Li et al., Cell, 2017). 

Publication (2017~2018):

  • Li, Y., Mathis A., Benjamin GF., Osterhout J., Ahanonu B.,Schnizter M.,Venkatesh N and Dulac C+ (2017). Neuronal representation of social information in the medial amygdala of behaving mice. Cell 171(5), 1176-1190.

Contact Information: yingli01 (at) fas.harvard.edu

Changliang Liu, Ph.D.

Department of Neurobiology, Harvard Medical School

Dr. Liu obtained his PhD from the Chinese Academy of Sciences in 2013 and has been working as a postdoc in the Department of Neurobiology at Harvard Medical School. His research is focused on molecular machinery (active zone) underlying neurotransmitter release and mechanism of dopamine signaling. He has recently identified the release sites of dopamine, which changed the long-standing dogma that dopamine transmission lacks endogenous architecture. Liu is also very interested in drug research. He has founded a biotech startup (https://cambriandiscovery.com/) aiming at developing cutting-edge new tools for drug discovery and development. 

Publication (2017~2018):

  • Changliang Liu, Lauren Kershberg, Jiexin Wang, Shirin Schneeberger, Pascal S. Kaeser, Dopamine secretion is mediated by sparse active zone-like release sites, Cell, 8 February 2018, Pages 706–718.e15 
  • Manyan Wong, Changliang Liu, Aram C.f. Roquas, Stephen Fowler, Pascal S, Kaeser, Liprin-α3 controls active zone composition, synaptic vesicle docking and exocytosis in hippocampal nerve terminals, PNAS, 2018 Feb 8. pii: 201719012 
  • Changliang Liu, Yakun Wang, Guozhang Jin, Weixing Shi, Ming Gao, Cocaine-induced locomotor sensitization associates with slow oscillatory firing of neurons in the ventral tegmental area, Sci Rep., 2018 Feb 19;8(1):3274 
  • Jiali Song, Juan Zhang, Changliang Liu, Chao Liu, Kongkai Zhu, Feifei Yang, Xigong Liu, João Paulo Figueiró Longo , Luis Alexandre Muehlmann, Ricardo Bentes Azevedo, Yu-Ying Zhang, Yuewei Guo, Cheng-Shi Jiang, Hua Zhang, Design and synthesis of pregnenolone/2-cyanoacryloyl conjugates with dual NF-κB inhibitory and anti-proliferative activities, Bioorg Med Chem Lett. 2017 Oct 15;27(20):4682-4686

Contact Information: Changliang_Liu (at) hms.harvard.edu

Nan Liu, Ph.D.

Boston Children’s Hospital, Harvard Medical School

Dr. Liu obtained his PhD in 2015 in National Institute of Biological Sciences, Beijing, where he studied the mechanism of heterchromatin spreading under the supervision of Dr. Bing Zhu. After PhD he decided to move on his study of epigenetic regulation to a defined developmental process, and he joined Dr. Stuart Orkin’s lab in 2017. Here he worked on how γ-globin is switched off during fetal to adult globin switch in blood cells. His recent finding provided a new model for this biological process, and unexpectedly, answered a longstanding question in the field raised several decades ago.

Publication (2017~2018):

  • Liu N, Hargreaves V, Zhu Q, Kurland JV, Hong J, Kim W, Sher F, Macias-Trevino C, Rogers JM, Kurita R, Nakamura Y, Yuan G-C, Bauer DE, Xu J, Bulyk ML, Orkin SH. Direct Promoter Repression by BCL11A Controls the Fetal to Adult Hemoglobin Switch. Cell 2018; 173:430-42.

Xiao (Shawn) Liu, Ph.D. 

Whitehead Institute for Biomedical Research, MIT

Dr. Liu was interested in understanding the molecular mechanisms of biological processes. His PhD study demonstrated how cell division was regulated by Polo-like kinase 1-mediated phosphorylation of diverse substrate proteins. As a postdoctoral fellow in Rudolf Jaenisch’s lab at Whitehead Institute, he developed a molecular tool for precise epigenome editing that enables the functional interrogation of individual epigenetic events. Using this tool, he demonstrated that demethylation of the hypermethylated CGG repeats at the pathological locus of FMR1 can restore FMR1 expression and rescue the electrophysiological abnormalities associated with Fragile X syndrome neurons. His career goal is to become an independent investigator in academia, and to uncover the epigenetic mechanisms that underlie brain functions and disorders at a cellular and molecular level.

Publication (2017~2018):

  • Liu, X.S., Wu, H., Krzisch, M., Wu, X., Muffat, J., Hnisz, D., Li, C., Yuan, B., Vershkov, D., Benvenisty, N., Young, R.A., and Jaenisch, R. (2018). Rescue of Fragile X syndrome by DNA methylation editing of the FMR1. Cell. 173, 1-14. 

Liang Tao, Ph.D.

Boston Children’s Hospital, Harvard Medical School

Dr. Tao received his Ph. D. degree in Biology from University of Science and Technology of China in 2010. Following a two-year postdoctoral research at Kansas University Medical Center, he joined Dr. Min Dong’s lab at Harvard Medical School in 2013. He has been investigating human pathogens and host-bacteria interactions, and mainly worked on bacterial toxins in Dr. Dong’s lab. One of his major findings is the identification of frizzled proteins as epithelial receptors for Clostridium difficile toxin B, which was published on Nature in 2016. Afterwards, by collaborating with a group in UC Irvine, they also defined the molecular basis of how this toxin recognizes its receptor frizzled (Science, 2018). Along the lines of basic research, he is also keen on translating the insights and knowledge of toxins toward developing new scientific tools as well as therapeutics. In early 2018, he was promoted to the junior faculty rank of instructor at HMS. He has accepted an offer as an assistant professor in Westlake Institute for Advanced Science, and will establish an independent lab later this year.

Publication (2017~2018):

  • Chen, P.*, Tao, L.*, Wang, T., Zhang, J., He, A., Lam, K., Liu, Z., He, X., Perry, K., Dong, M., Jin, R., (2018) Structural basis for recognition of frizzled proteins by Clostridium difficile toxin B. Science (In Press)  (* co-first authors)
  • Tao, L.*, Peng, L.*, Berntsson, RP-A., Park, S., Yu, F., Boone, C., Stenmark, P., Krupp, J., and Dong, M., (2017) Engineered botulinum neurotoxin B with improved efficacy for targeting human receptors. Nature Communications 8, 53. (* co-first authors)

Contact Information: liang.tao (at) childrens.harvard.edu

Jingyi Wu, Ph.D.

Broad Institute of MIT and Harvard, 
Mass General Hospital, Harvard Medical School

Dr. Wu earned her bachelor degree from China Agricultural University in 2012, and is conducting her Ph.D. research in the laboratory of Dr. Xie Wei in the Tsinghua University. Her thesis project is to study chromatin dynamics in early mammalian development. Using ATAC-seq and CARM, She and her colleagues have systematically revealed how open chromatin reprograms during human and mouse embryogenesis for the first time and identified the key transcription factors regulating this process. She is now doing her postdoc in Bradley Bernstein lab in Broad Institute and MGH to study the epigenetic regulation in cancer.

Publication (2017~2018):

  • Wu J*, Xu J*, Liu B* et al. & Xie W. Chromatin analysis in human early development reveals epigenetic transition during ZGA. Nature. (2018) In press (*: co-first author)

Xuhua Wang, Ph.D.

Boston Children’s Hospital, Harvard Medical School

Dr. Wang is a postdoctoral research fellow from Prof. Zhigang He’s Lab at Boston Children’s Hospital, Harvard Medical School, studying brain spinal circuit basis for motor control and developing gene and stem cell based regenerative therapy to cure spinal cord injury or stroke. His works have been recently published in Cell and Neuron. His findings have important impact on developing treatment strategies to cure spinal cord injury and stroke and studies in brain machine interface.

Publication (2017~2018):

  • Wang, X.*; Liu, Y.*; Li, X.*; Zhang, Z. *; Yang, H.; Zhang, Y.; Williams, P. R.; Alwahab, N. S. A.; Kapur, K.; Yu, B.; Ding, H.; Chen, M.; Gerfen, C. R.; Wang, K. H.; He, Z. “Deconstruction of corticospinal circuits for goal-directed motor skills”Cell, 2017, 171, 440- 445.  (*co-first author)
  • Liu, Y.*; Wang, X.*; Li, W.*; Zhang, Q.; Li, Y.; Zhang, Z.; Zhu, J.; Chen, B.; Williams, R. P.; Zhang, Y.; Yu, B.; Gu, X.; He, Z. “A sensitized IGF1 treatment restores corticospinal axon-dependent functions”, Neuron, 2017, 95, 817–833.  (*co-first author)

Jinfang Zhang, Ph.D.

Beth Israel Deaconess Medical Center, Harvard Medical School

Dr. Zhang earned his Ph.D. degree in the year of 2012 at Peking University in China. From 2013 to 2015, he received the postdoctoral research training at Beth Israel Deaconess Medical Center, Harvard Medical School. In 2015, he was promoted to the instructor at Harvard Medical School. In 2017, he received the NIH/NCI K99/R00 pathway to independence award. Currently, his research interest is exploring and developing novel effective antitumor strategies.

Publication (2017~2018):

  • Zhang J*, Bu X*, Wang H*, Zhu Y, Geng Y, Nihira NT, Tan Y, Ci Y, Wu F, Dai X, Guo J, Huang YH, Fan C, Ren S, Sun Y, Freeman GJ+, Sicinski P+, Wei W+. Cyclin D-CDK4 kinase destabilizes PD-L1 via Cul3SPOP to control cancer immune surveillance. Nature. 2018 Jan. 4; 553(7686): 91-95. (*co-first author) 
  • Wan L*, Xu K*, Wei Y*, Zhang J*, Han T, Fry C, Zhang Z, Wan Y, Huang L, Yuan M, Xia W, Chang W, Huang W, Liu C, Chang Y, Liu J, Wu Y, Jin V, Dai X, Guo J, Liu J, Jiang S, Li J, Asara JM, Brown M, Hung MC, Wei W. Phosphorylation of EZH2 by AMPK suppresses PRC2 methyltransferase activity and oncogenic function. Molecular Cell. 2018 Jan 18; 69(2): 279-291. (*co-first author)
  • Wu F, Dai X, Gan W, Wan L, Li M, Mitsiades N, Wei W, Ding Q+, Zhang J+. Prostate cancer-associated mutation in SPOP impairs its ability to target Cdc20 for poly-ubiquitination and degradation. Cancer Letters. 2017 Jan 28; 385: 207-214. (+co-corresponding author)

Contact Information: jinfangz13 (at) icloud.com

Yiming Zhou, Ph.D.

Brigham and Women’s Hospital, Harvard Medical School

Dr. Zhou is a postdoctoral fellow at Brigham and Women’s Hospital, Harvard Medical School, and Broad Institute of MIT and Harvard. His work focuses on the calcium signaling in kidney diseases. He identified a TRPC5 channel blocker as the first ion channel-targeted therapy for progressive proteinuric kidney disease. He is expanding his research goals into harnessing the power of genomics, and particularly single cell technologies, to inform our understanding of calcium signaling in kidney, so that he may pursue targeted, precision therapies targeting calcium signaling pathways. 

Publication (2017~2018):

  • Y Zhou*, Castonguay P*, et al. A small molecule inhibitor of TRPC5 ion channels suppresses progressive kidney disease in animal models. Science 2017, 8 (6368), 1332-1336. (*: co-first author)

Sponsored by:


哈佛医学院华人专家学者联合会

Website: http://www.hms-cssa.org

微信公众号: hmscssa

HMS-CSSA助手微信号:hmscssa2007

(添加好友后可由助手号邀请加入 HMS-CSSA 学术群 (需实名)、HMS-CSSA 生活群 (鼓励实名))