Network


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

Hotspot


Dive into the research topics where Zhuo Yu is active.

Publication


Featured researches published by Zhuo Yu.


Journal of Clinical Investigation | 2016

Sorting protein VPS33B regulates exosomal autocrine signaling to mediate hematopoiesis and leukemogenesis

Hao Gu; Chiqi Chen; Xiaoxin Hao; Conghui Wang; Xiaocui Zhang; Zhen Li; Hongfang Shao; Hongxiang Zeng; Zhuo Yu; Li Xie; Fangzhen Xia; Feifei Zhang; Xiaoye Liu; Yaping Zhang; Haishan Jiang; Jun Zhu; Jiangbo Wan; Chun Wang; Wei Weng; Jingjing Xie; Minfang Tao; Cheng Cheng Zhang; Junling Liu; Guo-Qiang Chen; Junke Zheng

Certain secretory proteins are known to be critical for maintaining the stemness of stem cells through autocrine signaling. However, the processes underlying the biogenesis, maturation, and secretion of these proteins remain largely unknown. Here we demonstrate that many secretory proteins produced by hematopoietic stem cells (HSCs) undergo exosomal maturation and release that is controlled by vacuolar protein sorting protein 33b (VPS33B). Deletion of VPS33B in either mouse or human HSCs resulted in impaired exosome maturation and secretion as well as loss of stemness. Additionally, VPS33B deficiency led to a dramatic delay in leukemogenesis. Exosomes purified from either conditioned medium or human plasma could partially rescue the defects of HSCs and leukemia-initiating cells (LICs). VPS33B co-existed in exosomes with GDI2, VPS16B, FLOT1, and other known exosome markers. Mechanistically, VPS33B interacted with the GDI2/RAB11A/RAB27A pathway to regulate the trafficking of secretory proteins as exosomes. These findings reveal an essential role for VPS33B in exosome pathways in HSCs and LICs. Moreover, they shed light on the understanding of vesicle trafficking in other stem cells and on the development of improved strategies for cancer treatment.


Cell & Bioscience | 2012

Oocyte-like cells induced from mouse spermatogonial stem cells

Lu Wang; Jinping Cao; Ping Ji; Di Zhang; Lianghong Ma; Martin Dym; Zhuo Yu; Lixin Feng

BackgroundDuring normal development primordial germ cells (PGCs) derived from the epiblast are the precursors of spermatogonia and oogonia. In culture, PGCs can be induced to dedifferentiate to pluripotent embryonic germ (EG) cells in the presence of various growth factors. Several recent studies have now demonstrated that spermatogonial stem cells (SSCs) can also revert back to pluripotency as embryonic stem (ES)-like cells under certain culture conditions. However, the potential dedifferentiation of SSCs into PGCs or the potential generation of oocytes from SSCs has not been demonstrated before.ResultsWe report that mouse male SSCs can be converted into oocyte-like cells in culture. These SSCs-derived oocytes (SSC-Oocs) were similar in size to normal mouse mature oocytes. They expressed oocyte-specific markers and gave rise to embryos through parthenogenesis. Interestingly, the Y- and X-linked testis-specific genes in these SSC-Oocs were significantly down-regulated or turned off, while oocyte-specific X-linked genes were activated. The gene expression profile appeared to switch to that of the oocyte across the X chromosome. Furthermore, these oocyte-like cells lost paternal imprinting but acquired maternal imprinting.ConclusionsOur data demonstrate that SSCs might maintain the potential to be reprogrammed into oocytes with corresponding epigenetic reversals. This study provides not only further evidence for the remarkable plasticity of SSCs but also a potential system for dissecting molecular and epigenetic regulations in germ cell fate determination and imprinting establishment during gametogenesis.


Cell & Bioscience | 2015

Hypoxic metabolism in human hematopoietic stem cells

Fatih Kocabas; Li Xie; Jingjing Xie; Zhuo Yu; Ralph J. DeBerardinis; Wataru Kimura; Suwannee Thet; Ahmed F. Elshamy; Hesham Abouellail; Shalini Muralidhar; Xiaoye Liu; Chiqi Chen; Hesham A. Sadek; Cheng Cheng Zhang; Junke Zheng

BackgroundAdult hematopoietic stem cells (HSCs) are maintained in a microenvironment, known as niche in the endosteal regions of the bone marrow. This stem cell niche with low oxygen tension requires HSCs to adopt a unique metabolic profile. We have recently demonstrated that mouse long-term hematopoietic stem cells (LT-HSCs) utilize glycolysis instead of mitochondrial oxidative phosphorylation as their main energy source. However, the metabolic phenotype of human hematopoietic progenitor and stem cells (HPSCs) remains unknown.ResultsWe show that HPSCs have a similar metabolic phenotype, as shown by high rates of glycolysis, and low rates of oxygen consumption. Fractionation of human mobilized peripheral blood cells based on their metabolic footprint shows that cells with a low mitochondrial potential are highly enriched for HPSCs. Remarkably, low MP cells had much better repopulation ability as compared to high MP cells. Moreover, similar to their murine counterparts, we show that Hif-1α is upregulated in human HPSCs, where it is transcriptionally regulated by Meis1. Finally, we show that Meis1 and its cofactors Pbx1 and HoxA9 play an important role in transcriptional activation of Hif-1α in a cooperative manner.ConclusionsThese findings highlight the unique metabolic properties of human HPSCs and the transcriptional network that regulates their metabolic phenotype.


Cell & Bioscience | 2015

PTEN signaling is required for the maintenance of spermatogonial stem cells in mouse, by regulating the expressions of PLZF and UTF1.

Wei Zhou; Hongfang Shao; Di Zhang; Jian Dong; Wei Cheng; Lu Wang; Yincheng Teng; Zhuo Yu

BackgroundPten plays a crucial role in the stem cell maintenance in a few organs. Pten defect also causes the premature oocytes and ovary aging. We and other groups have found that the phosphatidylinositol-3-OH kinase (PI3K)-Akt signaling regulates the proliferation and differentiation of spermatogonial stem cells (SSCs). PTEN functions as a negative regulator of the PI3K pathway. Thus, we thought that the fate of SSCs might be controlled by Pten.ResultsWe report that promyelocytic leukaemia zinc finger (PLZF) and undifferentiated embryonic cell transcription factor 1 (UTF1), both of which are germ cell-specific transcriptional factors, are regulated by Pten. Conditional deletion of Pten leads to reduction in PLZF expression but induction of UTF1, which is associated with SSCs depletion and infertility in males with age.ConclusionOur data demonstrate that Pten is required for the long-term maintenance of SSCs and precise regulation of spermatogenesis in mouse. The finding of a Pten-regulated GFRα1+/PLZF−/UTF1+ progenitor population provides a new insight into the precise mechanisms controlling SSC fate.


Journal of Hematology & Oncology | 2016

CD274 promotes cell cycle entry of leukemia-initiating cells through JNK/Cyclin D2 signaling

Xia Fang; Chiqi Chen; Fangzhen Xia; Zhuo Yu; Yaping Zhang; Feifei Zhang; Hao Gu; Jiangbo Wan; Xiaocui Zhang; Wei Weng; Cheng Cheng Zhang; Guo-Qiang Chen; Aibing Liang; Li Xie; Junke Zheng

BackgroundCD274 (programmed death ligand 1, also known as B7H1) is expressed in both solid tumors and hematologic malignancies and is of critical importance for the escape of tumor cells from immune surveillance by inhibiting T cell function via its receptor, programmed death 1 (PD-1). Increasing evidence indicates that functional monoclonal antibodies of CD274 may potently enhance the antitumor effect in many cancers. However, the role of CD274 in leukemia-initiating cells (LICs) remains largely unknown.MethodsWe established an MLL-AF9-induced acute myeloid leukemia (AML) model with wild-type (WT) and CD274-null mice to elucidate the role of CD274 in the cell fates of LICs, including self-renewal, differentiation, cell cycle, and apoptosis. RNA sequencing was performed to reveal the potential downstream targets, the results of which were further validated both in vitro and in vivo.ResultsIn silico analysis indicated that CD274 level was inversely correlated with the overall survival of AML patients. In Mac-1+/c-Kit+ mouse LICs, CD274 was expressed at a much higher level than in the normal hematopoietic stem cells (HSCs). The survival of the mice with CD274-null leukemia cells was dramatically extended during the serial transplantation compared with that of their WT counterparts. CD274 deletion led to a significant decrease in LIC frequency and arrest in the G1 phase of the cell cycle. Interestingly, CD274 is not required for the maintenance of HSC pool as shown in our previous study. Mechanistically, we demonstrated that the levels of both phospho-JNK and Cyclin D2 were strikingly downregulated in CD274-null LICs. The overexpression of Cyclin D2 fully rescued the loss of function of CD274. Moreover, CD274 was directly associated with JNK and enhanced the downstream signaling to increase the Cyclin D2 level, promoting leukemia development.ConclusionsThe surface immune molecule CD274 plays a critical role in the proliferation of LICs. The CD274/JNK/Cyclin D2 pathway promotes the cell cycle entry of LICs, which may serve as a novel therapeutic target for the treatment of leukemia.


Oncotarget | 2016

ChREBP promotes the differentiation of leukemia-initiating cells to inhibit leukemogenesis through the TXNIP/RUNX1 pathways

Hongxiang Zeng; Hao Gu; Chiqi Chen; Minle Li; Fangzhen Xia; Li Xie; Xiaoye Liu; Feifei Zhang; Xuemei Tong; Jiangbo Wang; Zhuo Yu; Junke Zheng

Targeting leukemia-initiating cells (LICs) is the key to eradicating leukemia and preventing its relapse. Recent studies have indicated that metabolic regulation may play a critical role in the maintenance of stemness in LICs, although the detailed mechanisms are poorly understood. Herein, we provide intriguing evidence showing that a glucose-responsive transcription factor, carbohydrate responsive element binding protein (ChREBP), served as a tumor suppressor rather than an oncogene, as previously described, to inhibit the development of acute myeloid leukemia by promoting the differentiation of LICs. Using an MLL-AF9-induced murine leukemia model, we demonstrated that the deletion of ChREBP resulted in the blockage of the differentiation of LICs and significantly reduced survival in ChREBP-null leukemic mice. However, ChREBP was not required for the normal repopulation abilities of hematopoietic stem cells. ChREBP promoted leukemia cell differentiation through the direct inhibition of RUNX1 or the transactivation of TXNIP to downregulate the RUNX1 level and ROS generation. Moreover, knockdown of ChREBP in human leukemia THP1 cells led to markedly enhanced proliferation and decreased differentiation upon PMA treatment. Collectively, we unraveled an unexpected role of ChREBP in leukemogenesis, which may provide valuable clues for developing novel metabolic strategies for leukemia treatment.


Haematologica | 2017

CD244 maintains the proliferation ability of leukemia initiating cells through SHP-2/p27kip1 signaling

Feifei Zhang; Xiaoye Liu; Chiqi Chen; Jun Zhu; Zhuo Yu; Jingjing Xie; Li Xie; Haitao Bai; Yaping Zhang; Xia Fang; Hao Gu; Chun Wang; Wei Weng; Cheng Cheng Zhang; Guo-Qiang Chen; Aibing Liang; Junke Zheng

Targeting leukemia initiating cells is considered to be an effective way to cure leukemia, for which it is critical to identify novel therapeutic targets. Herein, we demonstrate that CD244, which was initially reported as a key regulator for natural killer cells, is highly expressed on both mouse and human leukemia initiating cells. Upon CD244 knockdown, human leukemia cell lines and primary leukemia cells have markedly impaired proliferation abilities both in vitro and in vivo. Interestingly, the repopulation ability of both mouse and human hematopoietic stem cells is not impaired upon CD244 knockdown. Using an MLL-AF9-induced murine acute myeloid leukemia model, we show that leukemogenesis is dramatically delayed upon CD244 deletion, together with remarkably reduced Mac1+/c-Kit+ leukemia cells (enriched for leukemia initiating cells). Mechanistically, we reveal that CD244 is associated with c-Kit and p27 except for SHP-2 as previously reported. CD244 co-operates with c-Kit to activate SHP-2 signaling to dephosphorylate p27 and maintain its stability to promote leukemia development. Collectively, we provide intriguing evidence that the surface immune molecule CD244 plays an important role in the maintenance of stemness of leukemia initiating cells, but not in hematopoietic stem cells. CD244 may represent a novel therapeutic target for the treatment of acute myeloid leukemia.


International Journal of Oral and Maxillofacial Surgery | 2013

Dermatofibrosarcoma protuberans on the right neck with superior vena cava syndrome: Case report and literature review

H.S. Ong; T. Ji; L. Wang; Zhuo Yu; C. Zhang

Dermatofibrosarcoma protuberans (DFSP) is an uncommon dermal soft tissue tumour of intermediate malignancy. A 44-year-old man presented to the hospital with a large lesion on the right upper chest and neck. Despite eight previous surgical excisions, the tumour had continued to recur. Contrast-enhanced computed tomography showed recurrence of the tumour, associated with superior vena cava (SVC) syndrome. He declined radical surgical resection of the recurrent tumour, which may have required right upper limb amputation. Targeted therapy with sunitinib malate was therefore introduced. This case demonstrates the recurrent nature of DFSP and the association of this lesion on the upper chest/neck with SVC syndrome. Primary wide radical resection is essential for better local control and to avoid the development of SVC syndrome.


Journal of Clinical Investigation | 2018

JAM3 maintains leukemia-initiating cell self-renewal through LRP5/AKT/β-catenin/CCND1 signaling

Yaping Zhang; Fangzhen Xia; Xiaoye Liu; Zhuo Yu; Li Xie; Ligen Liu; Chiqi Chen; Haishan Jiang; Xiaoxin Hao; Xiaoxiao He; Feifei Zhang; Hao Gu; Jun Zhu; Haitao Bai; Cheng Cheng Zhang; Guo-Qiang Chen; Junke Zheng

Leukemia-initiating cells (LICs) are responsible for the initiation, development, and relapse of leukemia. The identification of novel therapeutic LIC targets is critical to curing leukemia. In this report, we reveal that junctional adhesion molecule 3 (JAM3) is highly enriched in both mouse and human LICs. Leukemogenesis is almost completely abrogated upon Jam3 deletion during serial transplantations in an MLL-AF9–induced murine acute myeloid leukemia model. In contrast, Jam3 deletion does not affect the functions of mouse hematopoietic stem cells. Moreover, knockdown of JAM3 leads to a dramatic decrease in the proliferation of both human leukemia cell lines and primary LICs. JAM3 directly associates with LRP5 to activate the downstream PDK1/AKT pathway, followed by the downregulation of GSK3&bgr; and activation of &bgr;-catenin/CCND1 signaling, to maintain the self-renewal ability and cell cycle entry of LICs. Thus, JAM3 may serve as a functional LIC marker and play an important role in the maintenance of LIC stemness through unexpected LRP5/PDK1/AKT/GSK3&bgr;/&bgr;-catenin/CCND1 signaling pathways but not via its canonical role in cell junctions and migration. JAM3 may be an ideal therapeutic target for the eradication of LICs without influencing normal hematopoiesis.


Cell Reports | 2018

PPM1K Regulates Hematopoiesis and Leukemogenesis through CDC20-Mediated Ubiquitination of MEIS1 and p21

Xiaoye Liu; Feifei Zhang; Yaping Zhang; Xie Li; Chiqi Chen; Meiyi Zhou; Zhuo Yu; Yunxia Liu; Yuzheng Zhao; Xiaoxin Hao; Yabin Tang; Liang Zhu; Ligen Liu; Li Xie; Hao Gu; Hongfang Shao; Fangzhen Xia; Chunrong Yin; Minfang Tao; Jingjing Xie; Cheng Cheng Zhang; Yi Yang; Haipeng Sun; Guo-Qiang Chen; Junke Zheng

In addition to acting as building blocks for biosynthesis, amino acids might serve as signaling regulators in various physiological and pathological processes. However, it remains unknown whether amino acid levels affect the activities of hematopoietic stem cells (HSCs). By using a genetically encoded fluorescent sensor of the intracellular levels of branched-chain amino acids (BCAAs), we could monitor the dynamics of BCAA metabolism in HSCs. A mitochondrial-targeted 2C-type Ser/Thr protein phosphatase (PPM1K) promotes the catabolism of BCAAs to maintain MEIS1 and p21 levels by decreasing the ubiquitination-mediated degradation controlled by the E3 ubiquitin ligase CDC20. PPM1K deficiency led to a notable decrease in MEIS1/p21 signaling to reduce the glycolysis and quiescence of HSCs, followed by a severe impairment in repopulation activities. Moreover, the deletion of Ppm1k dramatically extended survival in a murine leukemia model. These findings will enhance the current understanding of nutrient signaling in metabolism and function of stem cells.

Collaboration


Dive into the Zhuo Yu's collaboration.

Top Co-Authors

Avatar

Junke Zheng

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Chiqi Chen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Feifei Zhang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Li Xie

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Guo-Qiang Chen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Hao Gu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Xiaoye Liu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Fangzhen Xia

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Yaping Zhang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Cheng Cheng Zhang

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge