Network


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

Hotspot


Dive into the research topics where Huizhen Zhao is active.

Publication


Featured researches published by Huizhen Zhao.


PLOS ONE | 2012

SIRT1 activators suppress inflammatory responses through promotion of p65 deacetylation and inhibition of NF-κB activity.

Hongying Yang; Wei Zhang; Heng Pan; Heidi Galonek Feldser; Elden Lainez; Christine A. Miller; Stewart Leung; Zhong Zhong; Huizhen Zhao; Sharon Sweitzer; Thomas Considine; Thomas V. Riera; Vipin Suri; Brian S. White; James L. Ellis; George P. Vlasuk; Christine Loh

Chronic inflammation is a major contributing factor in the pathogenesis of many age-associated diseases. One central protein that regulates inflammation is NF-κB, the activity of which is modulated by post-translational modifications as well as by association with co-activator and co-repressor proteins. SIRT1, an NAD+-dependent protein deacetylase, has been shown to suppress NF-κB signaling through deacetylation of the p65 subunit of NF-κB resulting in the reduction of the inflammatory responses mediated by this transcription factor. The role of SIRT1 in the regulation of NF-κB provides the necessary validation for the development of pharmacological strategies for activating SIRT1 as an approach for the development of a new class of anti-inflammatory therapeutics. We report herein the development of a quantitative assay to assess compound effects on acetylated p65 protein in the cell. We demonstrate that small molecule activators of SIRT1 (STACs) enhance deacetylation of cellular p65 protein, which results in the suppression of TNFα-induced NF-κB transcriptional activation and reduction of LPS-stimulated TNFα secretion in a SIRT1-dependent manner. In an acute mouse model of LPS-induced inflammation, the STAC SRTCX1003 decreased the production of the proinflammatory cytokines TNFα and IL-12. Our studies indicate that increasing SIRT1-mediated NF-κB deacetylation using small molecule activating compounds is a novel approach to the development of a new class of therapeutic anti-inflammatory agents.


Biochemistry | 2011

A Tale of Two Subunits: How the Neomorphic R132H IDH1 Mutation Enhances Production of αHG

Beth Pietrak; Huizhen Zhao; Hongwei Qi; Chad Quinn; Enoch Gao; Joseph G. Boyer; Nestor O. Concha; Kristin K. Brown; Chaya Duraiswami; Richard Wooster; Sharon Sweitzer; Benjamin J. Schwartz

Heterozygously expressed single-point mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2, respectively) render these dimeric enzymes capable of producing the novel metabolite α-hydroxyglutarate (αHG). Accumulation of αHG is used as a biomarker for a number of cancer types, helping to identify tumors with similar IDH mutations. With IDH1, it has been shown that one role of the mutation is to increase the rate of conversion from αKG to αHG. To improve our understanding of the function of this mutation, we have detailed the kinetics of the normal (isocitrate to αKG) and neomorphic (αKG to αHG) reactions, as well as the coupled conversion of isocitrate to αHG. We find that the mutant IDH1 is very efficient in this coupled reaction, with the ability to form αHG from isocitrate and NADP(+). The wild type/wild type IDH1 is also able to catalyze this conversion, though it is much more sensitive to concentrations of isocitrate. This difference in behavior can be attributed to the competitive binding between isocitrate and αKG, which is made more favorable for αKG by the neomorphic mutation at arginine 132. Thus, each partial reaction in the heterodimer is functionally isolated from the other. To test whether there is a cooperative effect resulting from the two subunits being in a dimer, we selectively inactivated each subunit with a secondary mutation in the NADP/H binding site. We observed that the remaining, active subunit was unaffected in its associated activity, reinforcing the notion of each subunit being functionally independent. This was further demonstrated using a monomeric form of IDH from Azotobacter vinelandii, which can be shown to gain the same neomorphic reaction when a homologous mutation is introduced into that protein.


Nature Communications | 2015

Crystallographic structure of a small molecule SIRT1 activator-enzyme complex.

Han Dai; April Case; Thomas V. Riera; Thomas Considine; Jessica E. Lee; Yoshitomo Hamuro; Huizhen Zhao; Yong Jiang; Sharon Sweitzer; Beth Pietrak; Benjamin J. Schwartz; Charles A. Blum; Jeremy S. Disch; Richard Caldwell; Bruce G. Szczepankiewicz; Christopher Oalmann; Pui Yee Ng; Brian H. White; Rebecca L. Casaubon; Radha Narayan; Karsten Koppetsch; Francis Bourbonais; Bo Wu; Junfeng Wang; Dongming Qian; Fan Jiang; Cheney Mao; Minghui Wang; Erding Hu; Joseph Wu

SIRT1, the founding member of the mammalian family of seven NAD+-dependent sirtuins, is composed of 747 amino acids forming a catalytic domain and extended N- and C-terminal regions. We report the design and characterization of an engineered human SIRT1 construct (mini-hSIRT1) containing the minimal structural elements required for lysine deacetylation and catalytic activation by small molecule sirtuin-activating compounds (STACs). Using this construct, we solved the crystal structure of a mini-hSIRT1-STAC complex, which revealed the STAC-binding site within the N-terminal domain of hSIRT1. Together with hydrogen-deuterium exchange mass spectrometry (HDX-MS) and site-directed mutagenesis using full-length hSIRT1, these data establish a specific STAC-binding site and identify key intermolecular interactions with hSIRT1. The determination of the interface governing the binding of STACs with human SIRT1 facilitates greater understanding of STAC activation of this enzyme, which holds significant promise as a therapeutic target for multiple human diseases.


Biochemistry | 2013

Mutant IDH1 Enhances the Production of 2-Hydroxyglutarate Due to Its Kinetic Mechanism.

Alan R. Rendina; Beth Pietrak; Angela Smallwood; Huizhen Zhao; Hongwei Qi; Chad Quinn; Nicholas D. Adams; Nestor O. Concha; Chaya Duraiswami; Sara H. Thrall; Sharon Sweitzer; Benjamin J. Schwartz

The human, cytosolic enzyme isocitrate dehydrogenase 1 (IDH1) reversibly converts isocitrate to α-ketoglutarate (αKG). Cancer-associated somatic mutations in IDH1 result in a loss of this normal function but a gain in a new or neomorphic ability to convert αKG to the oncometabolite 2-hydroxyglutarate (2HG). To improve our understanding of the basis for this phenomenon, we have conducted a detailed kinetic study of wild-type IDH1 as well as the known 2HG-producing clinical R132H and G97D mutants and mechanistic Y139D and (newly described) G97N mutants. In the reductive direction of the normal reaction (αKG to isocitrate), dead-end inhibition studies suggest that wild-type IDH1 goes through a random sequential mechanism, similar to previous reports on related mammalian IDH enzymes. However, analogous experiments studying the reductive neomorphic reaction (αKG to 2HG) with the mutant forms of IDH1 are more consistent with an ordered sequential mechanism, with NADPH binding before αKG. This result was further confirmed by primary kinetic isotope effects for which saturating with αKG greatly reduced the observed isotope effect on (D)(V/K)NADPH. For the mutant IDH1 enzyme, the change in mechanism was consistently associated with reduced efficiencies in the use of αKG as a substrate and enhanced efficiencies using NADPH as a substrate. We propose that the sum of these kinetic changes allows the mutant IDH1 enzymes to reductively trap αKG directly into 2HG, rather than allowing it to react with carbon dioxide and form isocitrate, as occurs in the wild-type enzyme.


Cytotechnology | 2002

Rapid Expression of Recombinant Proteins in Modified CHO Cells Using the Baculovirus System

Luciano Ramos; Lisa A. Kopec; Sharon Sweitzer; James A. Fornwald; Huizhen Zhao; Paul R. McAllister; Dean E. McNulty; John J. Trill; James Kane

Baculovirus containing the mammalianCMV promoter, in place of the insect polyhedronpromoter (BacMam), has been used to transientlytransfect COS, CHO and CHOE1a (CHO cells expressing theE1a transcriptional activator). Using this system forthe expression of a cellular adhesion factor (SAF-3) Fcfusion protein in CHOE1a, we found that levels ofexpression were highest with a MOI of 100, 20mM sodiumbutyrate, at 34 °C. Production increased furtherif the cells were resuspended in fresh medium, about3 × 106 cells ml-1, prior to addition of the virus. These conditions were used to express 3 secretedproteins, SAF-3-Fc, CD40-hexa his and Asp 2-Fc, and, at2 to 6 days post infection, protein levels ranged from4 ug ml-1 to 25 ug ml-1. Based on these results, theBacMam system represents a viable technique forproducing protein at ug ml-1 levels in a relatively shortperiod of time.


Biochemical Journal | 2011

Biochemical characterization of human HIF hydroxylases using HIF protein substrates that contain all three hydroxylation sites

Melissa B. Pappalardi; Dean E. McNulty; John D. Martin; Kelly E. Fisher; Yong Jiang; Matthew C. Burns; Huizhen Zhao; Thau Ho; Sharon Sweitzer; Benjamin Schwartz; Roland S. Annan; Robert A. Copeland; Peter J. Tummino; Lusong Luo

The HIF (hypoxia-inducible factor) plays a central regulatory role in oxygen homoeostasis. HIF proteins are regulated by three Fe(II)- and α-KG (α-ketoglutarate)-dependent prolyl hydroxylase enzymes [PHD (prolyl hydroxylase domain) isoenzymes 1-3 or PHD1, PHD2 and PHD3] and one asparaginyl hydroxylase [FIH (factor inhibiting HIF)]. The prolyl hydroxylases control the abundance of HIF through oxygen-dependent hydroxylation of specific proline residues in HIF proteins, triggering subsequent ubiquitination and proteasomal degradation. FIH inhibits the HIF transcription activation through asparagine hydroxylation. Understanding the precise roles and regulation of these four Fe(II)- and α-KG-dependent hydroxylases is of great importance. In the present paper, we report the biochemical characterization of the first HIF protein substrates that contain the CODDD (C-terminal oxygen-dependent degradation domain), the NODDD (N-terminal oxygen-dependent degradation domain) and the CAD (C-terminal transactivation domain). Using LC-MS/MS (liquid chromatography-tandem MS) detection, we show that all three PHD isoenzymes have a strong preference for hydroxylation of the CODDD proline residue over the NODDD proline residue and the preference is observed for both HIF1α and HIF2α protein substrates. In addition, steady-state kinetic analyses show differential substrate selectivity for HIF and α-KG in reference to the three PHD isoforms and FIH.


Biochemistry | 2008

Biochemical Characterization of Human Prolyl Hydroxylase Domain Protein 2 Variants Associated with Erythrocytosis

Melissa B. Pappalardi; John D. Martin; Yong Jiang; Matthew C. Burns; Huizhen Zhao; Thau Ho; Sharon Sweitzer; Leng Lor; Benjamin J. Schwartz; Kevin J. Duffy; Richard R. Gontarek; Peter J. Tummino; Robert A. Copeland; Lusong Luo

Prolyl hydroxylase domain proteins (PHD isozymes 1-3) regulate levels of the alpha-subunit of the hypoxia inducible factor (HIF) through proline hydroxylation, earmarking HIFalpha for proteosome-mediated degradation. Under hypoxic conditions, HIF stabilization leads to enhanced transcription and regulation of a multitude of processes, including erythropoiesis. Herein, we examine the biochemical characterization of PHD2 variants, Arg371His and Pro317Arg, identified from patients with familial erythrocytosis. The variants display differential effects on catalytic rate and substrate binding, implying that partial inhibition or selective inhibition with regard to HIFalpha isoforms of PHD2 could result in the phenotype displayed by patients with familial erythrocytosis.


Journal of Medicinal Chemistry | 2015

Identification of Purines and 7-Deazapurines as Potent and Selective Type I Inhibitors of Troponin I-Interacting Kinase (TNNI3K).

Brian G. Lawhorn; Joanne Philp; Yongdong Zhao; Christopher Louer; Marlys Hammond; Mui Cheung; Harvey E. Fries; Alan P. Graves; Lisa M. Shewchuk; Liping Wang; Joshua E. Cottom; Hongwei Qi; Huizhen Zhao; Rachel Totoritis; Guofeng Zhang; Benjamin J. Schwartz; Hu Li; Sharon Sweitzer; Dennis Alan Holt; Gregory J. Gatto; Lara S. Kallander

A series of cardiac troponin I-interacting kinase (TNNI3K) inhibitors arising from 3-((9H-purin-6-yl)amino)-N-methyl-benzenesulfonamide (1) is disclosed along with fundamental structure-function relationships that delineate the role of each element of 1 for TNNI3K recognition. An X-ray structure of 1 bound to TNNI3K confirmed its Type I binding mode and is used to rationalize the structure-activity relationship and employed to design potent, selective, and orally bioavailable TNNI3K inhibitors. Identification of the 7-deazapurine heterocycle as a superior template (vs purine) and its elaboration by introduction of C4-benzenesulfonamide and C7- and C8-7-deazapurine substituents produced compounds with substantial improvements in potency (>1000-fold), general kinase selectivity (10-fold improvement), and pharmacokinetic properties (>10-fold increase in poDNAUC). Optimal members of the series have properties suitable for use in in vitro and in vivo experiments aimed at elucidating the role of TNNI3K in cardiac biology and serve as leads for developing novel heart failure medicines.


Biochemistry | 2012

On the Catalytic Mechanism of Human ATP Citrate Lyase

Fan Fan; Howard J. Williams; Joseph G. Boyer; Taylor L. Graham; Huizhen Zhao; Ruth Lehr; Hongwei Qi; Benjamin J. Schwartz; Frank M. Raushel; Thomas D. Meek

ATP citrate lyase (ACL) catalyzes an ATP-dependent biosynthetic reaction which produces acetyl-coenzyme A and oxaloacetate from citrate and coenzyme A (CoA). Studies were performed with recombinant human ACL to ascertain the nature of the catalytic phosphorylation that initiates the ACL reaction and the identity of the active site residues involved. Inactivation of ACL by treatment with diethylpyrocarbonate suggested the catalytic role of an active site histidine (i.e., His760), which was proposed to form a phosphohistidine species during catalysis. The pH-dependence of the pre-steady-state phosphorylation of ACL with [γ-(33)P]-ATP revealed an ionizable group with a pK(a) value of ~7.5, which must be unprotonated for the catalytic phosphorylation of ACL to occur. Mutagenesis of His760 to an alanine results in inactivation of the biosynthetic reaction of ACL, in good agreement with the involvement of a catalytic histidine. The nature of the formation of the phospho-ACL was further investigated by positional isotope exchange using [γ-(18)O(4)]-ATP. The β,γ-bridge to nonbridge positional isotope exchange rate of [γ-(18)O(4)]-ATP achieved its maximal rate of 14 s(-1) in the absence of citrate and CoA. This rate decreased to 5 s(-1) when citrate was added, and was found to be 10 s(-1) when both citrate and CoA were present. The rapid positional isotope exchange rates indicated the presence of one or more catalytically relevant, highly reversible phosphorylated intermediates. Steady-state measurements in the absence of citrate and CoA showed that MgADP was produced by both wild type and H760A forms of ACL, with rates at three magnitudes lower than that of k(cat) for the full biosynthetic reaction. The ATPase activity of ACL, along with the small yet significant positional isotope exchange rate observed in H760A mutant ACL (~150 fold less than wild type), collectively suggested the presence of a second, albeit unproductive, phosphoryl transfer in ACL. Mathematical analysis and computational simulation suggested that the desorption of MgADP at a rate of ~7 s(-1) was the rate-limiting step in the biosynthesis of AcCoA and oxaloacetate.


Biophysical Journal | 2014

Crystallographic Structure of a Small Molecule SIRT1 Activator/Enzyme Complex

Han Dai; Huizhen Zhao; Yong Jiang; Sharon Sweitzer; Beth Pietrak; Benjamin Schwartz; William Henry Miller; Erding Hu; James L. Ellis

Human SIRT1, the most studied member of the mammalian family of seven NAD+-dependent sirtuins, is composed of 747 amino acids with a catalytic core and extended N- and C-terminal regions. Herein we report the design and characterization of a human SIRT1 construct (mini-hSIRT1) containing the minimal structural elements required for lysine deacetylation and catalytic activation by sirtuin-activating compounds (STACs). Crystal structures of mini-hSIRT1/STAC complexes define the STAC binding site within the N-terminal domain, revealing key intra-molecular interactions. Site-directed mutagenesis of full-length human SIRT1 confirmed the importance of key residues in the STAC-binding domain. This structural information, together with proton-deuteron exchange mass spectrometry data, suggests a possible mechanism of STAC-mediated catalytic activation. The definitive determination of the interface governing the binding of STACs with human SIRT1 facilitates greater understanding of STAC activation of this enzyme which holds significant promise as a therapeutic target for multiple human diseases.

Collaboration


Dive into the Huizhen Zhao's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Beth Pietrak

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge