Network


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

Hotspot


Dive into the research topics where Ke Shuai is active.

Publication


Featured researches published by Ke Shuai.


Nature Reviews Immunology | 2003

Regulation of JAK–STAT signalling in the immune system

Ke Shuai; Bin Liu

The cytokine-activated Janus kinase (JAK)–signal transducer and activator of transcription (STAT) pathway has an important role in the control of immune responses. Dysregulation of JAK–STAT signalling is associated with various immune disorders. The signalling strength, kinetics and specificity of the JAK–STAT pathway are modulated at many levels by distinct regulatory proteins. Here, we review recent studies on the regulation of the JAK–STAT pathway that will enhance our ability to design rational therapeutic strategies for immune diseases.


Cell | 1994

Interferon activation of the transcription factor Stat91 involves dimerization through SH2-phosphotyrosyl peptide interactions

Ke Shuai; Curt M. Horvath; Linda H.Tsai Huang; Sajjad A. Qureshi; David Cowburn; James E. Darnell

Stat91 (a 91 kd protein that acts as a signal transducer and activator of transcription) is inactive in the cytoplasm of untreated cells but is activated by phosphorylation on tyrosine in response to a number of polypeptide ligands, including interferon alpha (IFN-alpha) and IFN-gamma. We report here that the inactive Stat91 in the cytoplasm of untreated cells is a monomer and that upon IFN-gamma-induced phosphorylation it forms a stable homodimer. Only the dimer is capable of binding to a specific DNA sequence directing transcription. Through dissociation and reassociation assays, we show that dimerization of Stat91 is mediated through SH2-phosphotyrosyl peptide interactions. Dimerization involving SH2 recognition of specific phosphotyrosyl peptides may well provide a prototype for interactions among family members of STAT proteins to form different transcription complexes.


Cell | 2001

Arginine Methylation of STAT1 Modulates IFNα/β-Induced Transcription

Kerri A. Mowen; Jie Tang; Wei Zhu; Brandon T. Schurter; Ke Shuai; Harvey R. Herschman; Michael David

Abstract Transcriptional induction by interferons requires the tyrosine and serine phosphorylation of STAT transcription factors. The N-terminal region is highly homologous among the STAT proteins and surrounds a completely conserved arginine residue. Here we demonstrate arginine methylation of STAT1 by the protein arginine methyl-transferase PRMT1 as a novel requirement for IFNα/β-induced transcription. Methyl-thioadenosine, a methyl-transferase inhibitor that accumulates in many transformed cells, inhibits STAT1-mediated IFN responses. This inhibition arises from impaired STAT1-DNA binding due to an increased association of the STAT inhibitor PIAS1 with phosphorylated STAT1 dimers in the absence of arginine methylation. Thus, arginine methylation of STAT1 is an additional posttranslational modification regulating transcription factor function, and alteration of arginine methylation might be responsible for the lack of interferon responsiveness observed in many malignancies.


Molecular and Cellular Biology | 2002

Identification of a Nuclear Stat1 Protein Tyrosine Phosphatase

Johanna ten Hoeve; María de Jesús Ibarra-Sánchez; Yubin Fu; Wei Zhu; Michel J. Tremblay; Michael David; Ke Shuai

ABSTRACT Upon interferon (IFN) stimulation, Stat1 becomes tyrosine phosphorylated and translocates into the nucleus, where it binds to DNA to activate transcription. The activity of Stat1 is dependent on tyrosine phosphorylation, and its inactivation in the nucleus is accomplished by a previously unknown protein tyrosine phosphatase (PTP). We have now purified a Stat1 PTP activity from HeLa cell nuclear extract and identified it as TC45, the nuclear isoform of the T-cell PTP (TC-PTP). TC45 can dephosphorylate Stat1 both in vitro and in vivo. Nuclear extracts lacking TC45 fail to dephosphorylate Stat1. Furthermore, the dephosphorylation of IFN-induced tyrosine-phosphorylated Stat1 is defective in TC-PTP-null mouse embryonic fibroblasts (MEFs) and primary thymocytes. Reconstitution of TC-PTP-null MEFs with TC45, but not the endoplasmic reticulum (ER)-associated isoform TC48, rescues the defect in Stat1 dephosphorylation. The dephosphorylation of Stat3, but not Stat5 or Stat6, is also affected in TC-PTP-null cells. Our results identify TC45 as a PTP responsible for the dephosphorylation of Stat1 in the nucleus.


Oncogene | 2000

Modulation of STAT signaling by STAT-interacting proteins.

Ke Shuai

STATs (signal transducer and activator of transcription) play important roles in numerous cellular processes including immune responses, cell growth and differentiation, cell survival and apoptosis, and oncogenesis. In contrast to many other cellular signaling cascades, the STAT pathway is direct: STATs bind to receptors at the cell surface and translocate into the nucleus where they function as transcription factors to trigger gene activation. However, STATs do not act alone. A number of proteins are found to be associated with STATs. These STAT-interacting proteins function to modulate STAT signaling at various steps and mediate the crosstalk of STATs with other cellular signaling pathways. This article reviews the roles of STAT-interacting proteins in the regulation of STAT signaling.


Nature Reviews Immunology | 2005

Regulation of gene-activation pathways by PIAS proteins in the immune system

Ke Shuai; Bin Liu

The protein inhibitor of activated STAT (PIAS) family of proteins has been proposed to regulate the activity of many transcription factors, including signal transducer and activator of transcription proteins (STATs), nuclear factor-κB, SMA- and MAD-related proteins (SMADs), and the tumour-suppressor protein p53. PIAS proteins regulate transcription through several mechanisms, including blocking the DNA-binding activity of transcription factors, recruiting transcriptional corepressors or co-activators, and promoting protein sumoylation. Recent genetic studies support an in vivo function for PIAS proteins in the regulation of innate immune responses. In this article, we review the current understanding of the molecular basis, specificity and physiological roles of PIAS proteins in the regulation of gene-activation pathways in the immune system.


Development | 2005

DNA methylation controls the timing of astrogliogenesis through regulation of JAK-STAT signaling

Guoping Fan; Keri Martinowich; Mark H. Chin; Fei He; Shaun D. Fouse; Leah Hutnick; Daisuke Hattori; Weihong Ge; Yin Shen; Hao Wu; Johanna ten Hoeve; Ke Shuai; Yi E. Sun

DNA methylation is a major epigenetic factor that has been postulated to regulate cell lineage differentiation. We report here that conditional gene deletion of the maintenance DNA methyltransferase I (Dnmt1) in neural progenitor cells (NPCs) results in DNA hypomethylation and precocious astroglial differentiation. The developmentally regulated demethylation of astrocyte marker genes as well as genes encoding the crucial components of the gliogenic JAK-STAT pathway is accelerated in Dnmt1–/– NPCs. Through a chromatin remodeling process, demethylation of genes in the JAK-STAT pathway leads to an enhanced activation of STATs, which in turn triggers astrocyte differentiation. Our study suggests that during the neurogenic period, DNA methylation inhibits not only astroglial marker genes but also genes that are essential for JAK-STAT signaling. Thus, demethylation of these two groups of genes and subsequent elevation of STAT activity are key mechanisms that control the timing and magnitude of astroglial differentiation.


The EMBO Journal | 2006

UBP43 is a novel regulator of interferon signaling independent of its ISG15 isopeptidase activity

Oxana A. Malakhova; Keun Il Kim; Jiann-Kae Luo; Weiguo Zou; K. G. Suresh Kumar; Serge Y. Fuchs; Ke Shuai; Dong-Er Zhang

Interferons (IFNs) regulate diverse cellular functions through activation of the Janus kinase–signal transducer and activator of transcription (JAK–STAT) pathway. Lack of Ubp43, an IFN‐inducible ISG15 deconjugating enzyme, leads to IFN hypersensitivity in ubp43−/− mice, suggesting an important function of Ubp43 in downregulation of IFN responses. Here, we show that Ubp43 negatively regulates IFN signaling independent of its isopeptidase activity towards ISG15. Ubp43 functions specifically for type I IFN signaling by downregulating the JAK–STAT pathway at the level of the IFN receptor. Using molecular, biochemical, and genetic approaches, we demonstrate that Ubp43 specifically binds to the IFNAR2 receptor subunit and inhibits the activity of receptor‐associated JAK1 by blocking the interaction between JAK and the IFN receptor. These data implicate Ubp43 as a novel in vivo inhibitor of signal transduction pathways that are specifically triggered by type I IFN.


Cell | 2008

Resolution of Sister Centromeres Requires RanBP2-Mediated SUMOylation of Topoisomerase IIα

Meelad M. Dawlaty; Liviu Malureanu; Karthik B. Jeganathan; Esther Kao; Claudio Sustmann; Samuel Tahk; Ke Shuai; Rudolf Grosschedl; Jan M. van Deursen

RanBP2 is a nucleoporin with SUMO E3 ligase activity that functions in both nucleocytoplasmic transport and mitosis. However, the biological relevance of RanBP2 and the in vivo targets of its E3 ligase activity are unknown. Here we show that animals with low amounts of RanBP2 develop severe aneuploidy in the absence of overt transport defects. The main chromosome segregation defect in cells from these mice is anaphase-bridge formation. Topoisomerase IIalpha (Topo IIalpha), which decatenates sister centromeres prior to anaphase onset to prevent bridges, fails to accumulate at inner centromeres when RanBP2 levels are low. We find that RanBP2 sumoylates Topo IIalpha in mitosis and that this modification is required for its proper localization to inner centromeres. Furthermore, mice with low amounts of RanBP2 are highly sensitive to tumor formation. Together, these data identify RanBP2 as a chromosomal instability gene that regulates Topo IIalpha by sumoylation and suppresses tumorigenesis.


Nature Immunology | 2004

PIAS1 selectively inhibits interferon-inducible genes and is important in innate immunity

Bin Liu; Sheldon Mink; Kelly A. Wong; Natalie Stein; Crescent Getman; Paul W. Dempsey; Hong Wu; Ke Shuai

Interferon (IFN) activates the signal transducer and activator of transcription (STAT) pathway to regulate immune responses. The protein inhibitor of activated STAT (PIAS) family has been suggested to negatively regulate STAT signaling. To understand the physiological function of PIAS1, we generated Pias1−/− mice. Using PIAS1-deficient cells, we show that PIAS1 selectively regulates a subset of IFN-γ- or IFN-β-inducible genes by interfering with the recruitment of STAT1 to the gene promoter. The antiviral activity of IFN-γ or IFN-β was consistently enhanced by Pias1 disruption. Pias1−/− mice showed increased protection against pathogenic infection. Our data indicate that PIAS1 is a physiologically important negative regulator of STAT1 and suggest that PIAS1 is critical for the IFN-γ- or IFN-β-mediated innate immune responses.

Collaboration


Dive into the Ke Shuai's collaboration.

Top Co-Authors

Avatar

Bin Liu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samuel Tahk

University of California

View shared research outputs
Top Co-Authors

Avatar

Jiayu Liao

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Randy Yang

University of California

View shared research outputs
Top Co-Authors

Avatar

Xiaoping Rao

University of California

View shared research outputs
Top Co-Authors

Avatar

Chan D. Chung

University of California

View shared research outputs
Top Co-Authors

Avatar

Guoping Fan

University of California

View shared research outputs
Top Co-Authors

Avatar

Hong Wu

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge