21 results on '"Zongli Li"'
Search Results
2. Practices for running a research-oriented shared cryo-EM facility
- Author
-
Richard M. Walsh, Megan L. Mayer, Christopher H. Sun, Shaun Rawson, Remya Nair, Sarah M. Sterling, and Zongli Li
- Subjects
Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Molecular Biology ,Biochemistry - Abstract
The Harvard Cryo-Electron Microscopy Center for Structural Biology, which was formed as a consortium between Harvard Medical School, Boston Children’s Hospital, Dana-Farber Cancer Institute, and Massachusetts General Hospital, serves both academic and commercial users in the greater Harvard community. The facility strives to optimize research productivity while training users to become expert electron microscopists. These two tasks may be at odds and require careful balance to keep research projects moving forward while still allowing trainees to develop independence and expertise. This article presents the model developed at Harvard Medical School for running a research-oriented cryo-EM facility. Being a research-oriented facility begins with training in cryo-sample preparation on a trainee’s own sample, ideally producing grids that can be screened and optimized on the Talos Arctica via multiple established pipelines. The first option, staff assisted screening, requires no user experience and a staff member provides instant feedback about the suitability of the sample for cryo-EM investigation and discusses potential strategies for sample optimization. Another option, rapid access, allows users short sessions to screen samples and introductory training for basic microscope operation. Once a sample reaches the stage where data collection is warranted, new users are trained on setting up data collection for themselves on either the Talos Arctica or Titan Krios microscope until independence is established. By providing incremental training and screening pipelines, the bottleneck of sample preparation can be overcome in parallel with developing skills as an electron microscopist. This approach allows for the development of expertise without hindering breakthroughs in key research areas.
- Published
- 2022
3. The structure of TRPC ion channels
- Author
-
Jian Li, Zongli Li, Xu Zhang, Xiaojing Song, Rui Liu, and Jin Zhang
- Subjects
0301 basic medicine ,Materials science ,Protein Conformation ,Physiology ,Cryo-electron microscopy ,viruses ,Amino Acid Motifs ,Porins ,macromolecular substances ,TRPC5 ,environment and public health ,TRPC4 ,TRPC6 ,Structure-Activity Relationship ,03 medical and health sciences ,Transient receptor potential channel ,Transient Receptor Potential Channels ,0302 clinical medicine ,TRPC3 ,Animals ,Humans ,Calcium Signaling ,Molecular Biology ,Ion channel ,TRPC ,Cryoelectron Microscopy ,Cell Biology ,diagnosis ,030104 developmental biology ,Biophysics ,Calcium ,030217 neurology & neurosurgery - Abstract
Briefly review the recent structural work of transient receptor potential canonical (TRPC) ion channels by using electron cryo-microscopy (cryo-EM). The high resolution structures of TRPC3, TRPC4, TRPC5 and TRPC6 are discussed.
- Published
- 2019
- Full Text
- View/download PDF
4. TRPC4 ion channel regulations by small-molecular inhibitors and calmodulin
- Author
-
Jingli Zhang and Zongli Li
- Subjects
0301 basic medicine ,Binding Sites ,Subfamily ,Calmodulin ,biology ,Physiology ,Chemistry ,Context (language use) ,Cell Biology ,TRPC4 ,Protein Structure, Secondary ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,biology.protein ,Animals ,Humans ,Molecular Biology ,030217 neurology & neurosurgery ,Ion channel ,TRPC ,TRPC Cation Channels - Abstract
TRPC4 ion channel was reported to be regulated by small molecular inhibitors and calmodulin. We discuss these findings in the context of other members of TRPC subfamily modulated by different stimulants.
- Published
- 2021
- Full Text
- View/download PDF
5. 6S RNA Mimics B-Form DNA to Regulate Escherichia coli RNA Polymerase
- Author
-
Katherine Leon, Karen M. Wassarman, Thomas Walz, Jared T. Winkelman, Zongli Li, Shili Feng, Andrey Feklistov, Seth A. Darst, James Chen, and Elizabeth A. Campbell
- Subjects
0301 basic medicine ,DNA, Bacterial ,RNA, Untranslated ,RNA-dependent RNA polymerase ,Sigma Factor ,Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Transcription (biology) ,RNA polymerase ,RNA polymerase I ,Escherichia coli ,Molecular Biology ,RNA polymerase II holoenzyme ,Escherichia coli Proteins ,RNA ,Cell Biology ,DNA-Directed RNA Polymerases ,Non-coding RNA ,Molecular biology ,Cell biology ,RNA silencing ,RNA, Bacterial ,030104 developmental biology ,chemistry ,DNA, B-Form - Abstract
Summary Noncoding RNAs (ncRNAs) regulate gene expression in all organisms. Bacterial 6S RNAs globally regulate transcription by binding RNA polymerase (RNAP) holoenzyme and competing with promoter DNA. Escherichia coli ( Eco ) 6S RNA interacts specifically with the housekeeping σ 70 -holoenzyme (Eσ 70 ) and plays a key role in the transcriptional reprogramming upon shifts between exponential and stationary phase. Inhibition is relieved upon 6S RNA-templated RNA synthesis. We report here the 3.8 A resolution structure of a complex between 6S RNA and Eσ 70 determined by single-particle cryo-electron microscopy and validation of the structure using footprinting and crosslinking approaches. Duplex RNA segments have A-form C3′ endo sugar puckers but widened major groove widths, giving the RNA an overall architecture that mimics B-form promoter DNA. Our results help explain the specificity of Eco 6S RNA for Eσ 70 and show how an ncRNA can mimic B-form DNA to directly regulate transcription by the DNA-dependent RNAP.
- Published
- 2017
6. Age-related activation of MKK/p38/NF-κB signaling pathway in lung: From mouse to human
- Author
-
Yan Li, Zongli Li, Chen Wang, Wei Wang, Junfa Li, Junmin Huang, Huadong Du, Song Han, Kewu Huang, Xiaoxia Ren, and Xiujuan Yao
- Subjects
Adult ,Lipopolysaccharides ,Male ,Aging ,medicine.medical_specialty ,Lipopolysaccharide ,MAP Kinase Signaling System ,p38 mitogen-activated protein kinases ,medicine.medical_treatment ,Biochemistry ,chemistry.chemical_compound ,Endocrinology ,Stress, Physiological ,In vivo ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,Interleukin 6 ,Lung ,Molecular Biology ,Aged ,biology ,business.industry ,NF-kappa B ,Cell Biology ,Middle Aged ,beta-Galactosidase ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cytokine ,chemistry ,Immunology ,biology.protein ,Cytokines ,Tumor necrosis factor alpha ,business ,Ex vivo - Abstract
We and others previously reported that the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and IL-6 significantly accumulate with age in mouse lung. This is accompanied by elevated phosphorylation of p38. Here, we further investigate whether aging affects activation of p38 signaling and the inflammatory reaction after exposure to lipopolysaccharide (LPS) in the lungs of mice in vivo and humans ex vivo. The data showed that activation of p38 peaked at 0.5 h and then rapidly declined in young (2-month-old) mouse lung, after intranasal inhalation challenge with LPS. In contract, activation of p38 peaked at 24 h and was sustained longer in aged (20-month-old) mice. As well as altered p38, activations of its upstream activator MKK and downstream substrate NF-κB were also changed in the lungs of aged mice, which corresponded with the absence in the early phase but delayed increases in concentrations of TNF-α, IL-1β and IL-6. Consistent with the above observations in mice, similar patterns of p38 signaling also occurred in human lungs. Compared with younger lungs from adult–middle aged subjects, the activation of p38, MKK and NF-κB, as well as the production of pro-inflammatory cytokines were significantly increased in the lungs of older subjects ex vivo. Exposure of human lung cells to LPS induced rapid activation of p38, MKK and NF-κB in these cells from adult–middle aged subjects, but not older subjects, with increases in the production of the pro-inflammatory cytokines. The LPS-induced rapid activation in the lung cells from adult–middle aged subjects occurred as early as 0.25 h after exposure, and then declined. Compared with adult–middle aged subjects, the LPS exposure did not induce marked changes in the early phase, either in the activation of p38, MKK and NF-κB, or in the production of TNF-α, IL-1β or IL-6 in the lung cells from older subjects. In contrast, these changes occurred relatively late, peaked at 16 h and were sustained longer in the lungs of older subjects. These data support the hypothesis that the sustained activation of the p38 signaling pathway at baseline and the absence in the early phase but delayed of p38 signaling pathway response to LPS in the elderly may play important roles in increased susceptibility of aged lungs to inflammatory injury.
- Published
- 2014
- Full Text
- View/download PDF
7. Molecular Imprinting as a Signal-Activation Mechanism of the Viral RNA Sensor RIG-I
- Author
-
Thomas Walz, Bin Wu, Zongli Li, Pawel A. Penczek, Edward H. Egelman, Katharine E. Magor, Sun Hur, Alys Peisley, and David G. Tetrault
- Subjects
Models, Molecular ,Protein Conformation ,viruses ,chemical and pharmacologic phenomena ,Biology ,DEAD-box RNA Helicases ,Molecular Imprinting ,Protein filament ,Protein structure ,Viral rna ,Protein Structure, Quaternary ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Genetics ,Binding Sites ,RIG-I ,virus diseases ,Signal transducing adaptor protein ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,Protein Structure, Tertiary ,Microscopy, Electron ,Biophysics ,CARD domain ,RNA, Viral ,biological phenomena, cell phenomena, and immunity ,Molecular imprinting ,Adaptor molecule - Abstract
RIG-I activates interferon signaling pathways by promoting filament formation of the adaptor molecule, MAVS. Assembly of the MAVS filament is mediated by its CARD domain (CARD(MAVS)), and requires its interaction with the tandem CARDs of RIG-I (2CARD(RIG-I)). However, the precise nature of the interaction between 2CARD(RIG-I) and CARD(MAVS), and how this interaction leads to CARD(MAVS) filament assembly, has been unclear. Here we report a 3.6 Å electron microscopy structure of the CARD(MAVS) filament and a 3.4 Å crystal structure of the 2CARD(RIG-I):CARD(MAVS) complex, representing 2CARD(RIG-I) "caught in the act" of nucleating the CARD(MAVS) filament. These structures, together with functional analyses, show that 2CARD(RIG-I) acts as a template for the CARD(MAVS) filament assembly, by forming a helical tetrameric structure and recruiting CARD(MAVS) along its helical trajectory. Our work thus reveals that signal activation by RIG-I occurs by imprinting its helical assembly architecture on MAVS, a previously uncharacterized mechanism of signal transmission.
- Published
- 2014
- Full Text
- View/download PDF
8. Cryo-EM Structure of Caspase-8 Tandem DED Filament Reveals Assembly and Regulation Mechanisms of the Death Inducing Signaling Complex
- Author
-
Frank DiMaio, Katryn J. Stacey, Yang Li, Parimala R. Vajjhala, Tian-Min Fu, Richard M. Siegel, Devendra B. Srivastava, Zongli Li, Hao Wu, Edward H. Egelman, Alvin Lu, Pawel A. Penczek, and Anthony C. Cruz
- Subjects
0301 basic medicine ,Protein Conformation, alpha-Helical ,Death Domain Receptor Signaling Adaptor Proteins ,Cryo-electron microscopy ,Fas-Associated Death Domain Protein ,Recombinant Fusion Proteins ,CASP8 and FADD-Like Apoptosis Regulating Protein ,Gene Expression ,Apoptosis ,Caspase 8 ,Transfection ,Article ,Protein filament ,TNF-Related Apoptosis-Inducing Ligand ,03 medical and health sciences ,Jurkat Cells ,Viral Proteins ,medicine ,Humans ,Protein Interaction Domains and Motifs ,FADD ,Amino Acid Sequence ,fas Receptor ,Molecular Biology ,Binding Sites ,030102 biochemistry & molecular biology ,biology ,Sequence Homology, Amino Acid ,Death Effector Domain ,Mutagenesis ,Cryoelectron Microscopy ,Inflammasome ,Cell Biology ,Cell biology ,CARD Signaling Adaptor Proteins ,Cytoskeletal Proteins ,030104 developmental biology ,Death-inducing signaling complex ,biology.protein ,Death effector domain ,Protein Conformation, beta-Strand ,Sequence Alignment ,medicine.drug ,Plasmids ,Protein Binding - Abstract
Caspase-8 activation can be triggered by death receptor-mediated formation of the death-inducing signaling complex (DISC) and by the inflammasome adaptor ASC. Caspase-8 assembles with FADD at the DISC and with ASC at the inflammasome through its tandem death effector domain (tDED), which is regulated by the tDED-containing cellular inhibitor cFLIP and the viral inhibitor MC159. Here we present the caspase-8 tDED filament structure determined by cryoelectron microscopy. Extensive assembly interfaces not predicted by the previously proposed linear DED chain model were uncovered, and were further confirmed by structure-based mutagenesis in filament formation in vitro and Fas-induced apoptosis and ASC-mediated caspase-8 recruitment in cells. Structurally, the two DEDs in caspase-8 use quasi-equivalent contacts to enable assembly. Using the tDED filament structure as a template, structural analyses reveal the interaction surfaces between FADD and caspase-8 and the distinct mechanisms of regulation by cFLIP and MC159 through comingling and capping, respectively.
- Published
- 2016
9. Structural basis for allosteric regulation of human ribonucleotide reductase by nucleotide-induced oligomerization
- Author
-
Shalini Jha, Thomas Walz, A.K. Roos, R Martin Welin, Faiz Ahmad, P. Nordlund, Hai Xu, Sanath R. Wijerathna, Zongli Li, Ryo Nakano, Chris Dealwis, Jay S. Prendergast, James W. Fairman, and Susanne Flodin
- Subjects
Models, Molecular ,Protein subunit ,Allosteric regulation ,Saccharomyces cerevisiae ,Random hexamer ,Crystallography, X-Ray ,Article ,Deoxyadenine Nucleotides ,Allosteric Regulation ,Structural Biology ,Oxidoreductase ,Catalytic Domain ,Ribonucleotide Reductases ,Humans ,heterocyclic compounds ,Nucleotide ,Molecular Biology ,chemistry.chemical_classification ,biology ,Nucleotides ,Activator (genetics) ,Chemistry ,biology.organism_classification ,enzymes and coenzymes (carbohydrates) ,Ribonucleotide reductase ,Biochemistry ,Mutagenesis, Site-Directed ,Protein Multimerization - Abstract
Ribonucleotide reductase (RR) is an αnβn (RR1●RR2) complex that maintains balanced dNTP pools by reducing ribonucleoside diphosphates to deoxyribonucleoside diphosphates. RR1 is the catalytic subunit and RR2 houses the free radical required for catalysis. RR is allosterically regulated by its activator ATP and its inhibitor dATP, which regulate RR activity by inducing oligomerization of RR1. Here, we report the first X-ray structures of human RR1 bound to TTP-only, dATP-only, TTP●GDP, TTP●ATP, and TTP●dATP. These structures provide insights into ATP/dATP regulation of RR. At physiological dATP concentrations, RR1 forms inactive hexamers. We determined the first X-ray structure of the RR1●dATP hexamer and used single-particle electron microscopy to visualize the α6●ββ’ 1●dATP holo complex. Site-directed mutagenesis and functional assays confirm that hexamerization is a prerequisite for inhibition by dATP. Our data provide an elegant mechanism for regulating RR activity by dATP-induced oligomerization.
- Published
- 2011
- Full Text
- View/download PDF
10. Structure of the influenza virus A H5N1 nucleoprotein: implications for RNA binding, oligomerization, and vaccine design
- Author
-
Andy Ka-Leung Ng, Sui-Mui Li, Paul K.S. Chan, Jia-huai Wang, Pang-Chui Shaw, Wood Yee Chan, Kemin Tan, Jin-huan Liu, Hongmin Zhang, Andrzej Joachimiak, Shannon Wing-Ngor Au, Zongli Li, and Thomas Walz
- Subjects
Protein Conformation ,Viral protein ,Molecular Sequence Data ,RNA-binding protein ,Biology ,Crystallography, X-Ray ,medicine.disease_cause ,Antiviral Agents ,Biochemistry ,Epitope ,Virus ,Research Communications ,Protein structure ,Genetics ,Influenza A virus ,medicine ,Amino Acid Sequence ,Molecular Biology ,Influenza A Virus, H5N1 Subtype ,Viral Core Proteins ,RNA ,Virology ,Nucleoprotein ,Nucleoproteins ,Influenza Vaccines ,Drug Design ,Biophysics ,Biotechnology - Abstract
The threat of a pandemic outbreak of influenza virus A H5N1 has become a major concern worldwide. The nucleoprotein (NP) of the virus binds the RNA genome and acts as a key adaptor between the virus and the host cell. It, therefore, plays an important structural and functional role and represents an attractive drug target. Here, we report the 3.3-Å crystal structure of H5N1 NP, which is composed of a head domain, a body domain, and a tail loop. Our structure resolves the important linker segments (residues 397–401, 429–437) that connect the tail loop with the remainder of the molecule and a flexible, basic loop (residues 73–91) located in an arginine-rich groove surrounding Arg150. Using surface plasmon resonance, we found the basic loop and arginine-rich groove, but mostly a protruding element containing Arg174 and Arg175, to be important in RNA binding by NP. We also used our crystal structure to build a ring-shaped assembly of nine NP subunits to model the miniribonucleoprotein particle previously visualized by electron microscopy. Our study of H5N1 NP provides insight into the oligomerization interface and the RNA-binding groove, which are attractive drug targets, and it identifies the epitopes that might be used for universal vaccine development.—Ng, A. K.-L., Zhang, H., Tan, K., Li, Z., Liu, J.-h., Chan, P. K.-S., Li, S.-M., Chan, W.-Y., Au, S. W.-N., Joachimiak, A., Walz, T., Wang, J.-H., Shaw, P.-C. Structure of the influenza virus A H5N1 nucleoprotein: implications for RNA binding, oligomerization, and vaccine design.
- Published
- 2008
- Full Text
- View/download PDF
11. Transglutaminase Induces Protofibril-like Amyloid β-Protein Assemblies That Are Protease-resistant and Inhibit Long-term Potentiation
- Author
-
Thomas Walz, Dean M. Hartley, Gavitt A. Woodard, Shaomin Li, Austin C. Speier, Chaohui Zhao, and Zongli Li
- Subjects
Proteases ,Amyloid ,Tissue transglutaminase ,Guinea Pigs ,Long-Term Potentiation ,Hippocampus ,Insulysin ,Biochemistry ,Mice ,Alzheimer Disease ,GTP-Binding Proteins ,Insulin-degrading enzyme ,medicine ,Animals ,Humans ,Protein Glutamine gamma Glutamyltransferase 2 ,Molecular Biology ,Neprilysin ,Amyloid beta-Peptides ,Transglutaminases ,integumentary system ,biology ,Neurodegeneration ,Long-term potentiation ,Cell Biology ,medicine.disease ,Cross-Linking Reagents ,Protein Structure and Folding ,Disease Progression ,Biophysics ,biology.protein ,Peptides ,Peptide Hydrolases - Abstract
An increasing body of evidence suggests that soluble assemblies of amyloid beta-protein (Abeta) play an important role in the initiation of Alzheimer disease (AD). In vitro studies have found that synthetic Abeta can form soluble aggregates through self-assembly, but this process requires Abeta concentrations 100- to 1000-fold greater than physiological levels. Tissue transglutaminase (TGase) has been implicated in neurodegeneration and can cross-link Abeta. Here we show that TGase induces rapid aggregation of Abeta within 0.5-30 min, which was not observed with chemical cross-linkers. Both Abeta40 and Abeta42 are good substrates for TGase but show different aggregation patterns. Guinea pig and human TGase induced similar Abeta aggregation patterns, and oligomerization was observed with Abeta40 concentrations as low as 50 nm. The formed Abeta40 species range from 5 to 6 nm spheres to curvilinear structures of the same width, but up to 100 nm in length, that resemble the previously described self-assembled Abeta protofibrils. TGase-induced Abeta40 assemblies are resistant to a 1-h incubation with either neprilysin or insulin degrading enzyme, whereas the monomer is rapidly degraded by both proteases. In support of these species being pathological, TGase-induced Abeta40 assemblies (100 nm) inhibited long term potentiation recorded in the CA1 region of mouse hippocampus slices. Our data suggest that TGase can contribute to AD by initiating Abeta oligomerization and aggregation at physiological levels, by reducing the clearance of Abeta due to the generation of protease-resistant Abeta species, and by forming Abeta assemblies that inhibit processes involved in memory and learning. Our data suggest that TGase might constitute a specific therapeutic target for slowing or blocking the progression of AD.
- Published
- 2008
- Full Text
- View/download PDF
12. Coat protein fold and maturation transition of bacteriophage P22 seen at subnanometer resolutions
- Author
-
Wah Chiu, Zhixian Zhang, Peter E. Prevelige, Matthew L. Baker, Wen Jiang, and Zongli Li
- Subjects
Models, Molecular ,Protein Folding ,Conformational change ,Transition (genetics) ,Fold (higher-order function) ,biology ,Protein Conformation ,viruses ,Cryoelectron Microscopy ,Capsomere ,biochemical phenomena, metabolism, and nutrition ,Coat protein ,Protein complex assembly ,biology.organism_classification ,Biochemistry ,Molecular biology ,Protein Structure, Secondary ,Bacteriophage ,Protein Subunits ,Capsid ,Structural Biology ,Genetics ,Biophysics ,Capsid Proteins ,Bacteriophage P22 - Abstract
Bacteriophage P22 is a prototypical biological machine used for studying protein complex assembly and capsid maturation. Using cryo-EM, we solved the structures of P22 before and after the capsid maturation at 8.5 A and 9.5 A resolutions, respectively. These structures allowed visualization of alpha-helices and beta-sheets from which the capsid protein fold is derived. The capsid fold is similar to that of the coat protein of HK97 bacteriophage. The cryo-EM shows that a large conformational change of the P22 capsid during maturation transition involves not only the domain movement of individual subunits, but also refolding of the capsid protein.
- Published
- 2003
- Full Text
- View/download PDF
13. Altered Th1/Th2 commitment contributes to lung senescence in CXCR3-deficient mice
- Author
-
Xiujuan Yao, Junmin Huang, Xiaoxia Reng, Yan Li, Zongli Li, Clarke G. Tankersley, Kewu Huang, Jinming Gao, Junfa Li, Wei Wang, and Chen Wang
- Subjects
Senescence ,Male ,Vital capacity ,medicine.medical_specialty ,Chemokine ,Aging ,Receptors, CXCR3 ,medicine.medical_treatment ,Population ,Cell Count ,Biochemistry ,Mice ,Endocrinology ,Th2 Cells ,Internal medicine ,Genetics ,medicine ,Animals ,Lung volumes ,education ,Molecular Biology ,Lung ,Th1-Th2 Balance ,Mice, Knockout ,education.field_of_study ,biology ,Cell Biology ,Immunosenescence ,Organ Size ,respiratory system ,Th1 Cells ,Mice, Inbred C57BL ,Cytokine ,medicine.anatomical_structure ,Immunology ,Models, Animal ,biology.protein ,Cytokines ,Collagen ,Chemokines - Abstract
Aging is an inevitable process associated with immune imbalance, which is characterized by a progressive functional decline in major organs, including lung. However, effects of altered Th1/Th2 commitment on lung senescence are largely unknown. To examine effects of altered Th1/Th2 balance on lung aging, we measured proportions of Th1 and Th2 cells and expression of cytokines, chemokines, collagen deposition and other relevant physiological and pathological parameters in 2- and 20-months-old (mo) CXCR3-deficient (CXCR3(-/-)) C57BL/6J mice compared with wild-type (WT) mice. There was a significant weight-loss observed in 20-mo CXCR3(-/-) mice compared with the same aged WT group. Although lung function and structure changed with age in both groups, central airway resistance (Rn), tissue elastance (H) and damping (G) were significantly lower in 20-mo CXCR3(-/-) mice than those of WT mice. In contrast, the whole lung volume (V(L)), the mean linear intercept length of alveolar (L(m)), and the total lung collagen content were significantly elevated in 20-mo CXCR3(-/-) mice. With aging, the lungs of WT mice had typical Th1-type status (increased population of Th1 cells and concentrations of cytokine IFN-γ and CXCR3 ligands) while CXCR3(-/-) mice showed Th2-type polarization (decreased proportion of Th1 cells and concentrations of CXCR3 ligands but increased level of IL-4). Our data suggest that Immunosenescence is associated with lung aging, and that altered Th1/Th2 imbalance favors Th2 predominance in CXCR3(-/-) mice, which contributes to the process of accelerated lung aging in this model.
- Published
- 2012
14. Simultaneous visualization of the extracellular and cytoplasmic domains of the epidermal growth factor receptor
- Author
-
Li-Zhi Mi, Thomas Walz, Chafen Lu, Noritaka Nishida, Timothy A. Springer, and Zongli Li
- Subjects
Conformational change ,biology ,Chemistry ,Kinase ,Active site ,Ligands ,Article ,Protein Structure, Tertiary ,ErbB Receptors ,Transmembrane domain ,Protein structure ,Biochemistry ,Ectodomain ,Structural Biology ,Catalytic Domain ,Mutation ,biology.protein ,Biophysics ,Humans ,Epidermal growth factor receptor ,Receptor ,Molecular Biology ,Dimerization - Abstract
To our knowledge, no structural study to date has characterized, in an intact receptor, the coupling of conformational change in extracellular domains through a single-pass transmembrane domain to conformational change in cytoplasmic domains. Here we examine such coupling, and its unexpected complexity, using nearly full-length epidermal growth factor receptor (EGFR) and negative-stain EM. The liganded, dimeric EGFR ectodomain can couple both to putatively active, asymmetrically associated kinase dimers and to putatively inactive, symmetrically associated kinase dimers and monomers. Inhibitors that stabilize the active or inactive conformation of the kinase active site, as well as mutations in the kinase dimer interface and a juxtamembrane phosphorylation site, shift the equilibrium among the three kinase association states. This coupling of one conformation of an activated receptor ectodomain to multiple kinase-domain arrangements reveals previously unanticipated complexity in transmembrane signaling and facilitates regulation of receptor function in the juxtamembrane and cytoplasmic environments.
- Published
- 2011
15. Age-induced augmentation of p38 MAPK phosphorylation in mouse lung
- Author
-
Xu Liu, Xiangning Bu, Zongli Li, Chen Wang, Clarke G. Tankersley, Kewu Huang, and Junfa Li
- Subjects
MAPK/ERK pathway ,Male ,medicine.medical_specialty ,Aging ,p38 mitogen-activated protein kinases ,Blotting, Western ,Interleukin-1beta ,Inflammation ,Enzyme-Linked Immunosorbent Assay ,Biochemistry ,p38 Mitogen-Activated Protein Kinases ,Oxidative Phosphorylation ,Superoxide dismutase ,Mice ,Endocrinology ,Internal medicine ,Genetics ,medicine ,Animals ,Protein kinase A ,Molecular Biology ,Lung ,chemistry.chemical_classification ,Reactive oxygen species ,medicine.diagnostic_test ,biology ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Cell Biology ,Immunohistochemistry ,Bronchoalveolar lavage ,chemistry ,Immunology ,biology.protein ,Cytokines ,Tumor necrosis factor alpha ,medicine.symptom ,Reactive Oxygen Species - Abstract
The p38 mitogen-activated protein kinase (p38 MAPK) pathway is a key regulator of pro-inflammatory cytokine biosynthesis, which may contribute to the chronic low-grade inflammation observed with aging. We hypothesize that aging up-regulates the activation of p38 MAPK as well as the pro-inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) in mouse lung, and is accompanied by disturbances in oxidant-antioxidant status. In addition, the elevated protein levels of phosphorylated active form of p38 MAPK (phospho-p38 MAPK) with age are tissue-specific. To test this hypothesis, protein levels of phospho-p38 MAPK were determined using Western blot analysis in isolated lung, brain, heart, spleen, kidney and muscle of young (2-month-old) and aged (20-month-old) male C57BL/6J mice. Results show that phospho-p38 MAPK protein levels, not total-p38 MAPK, increased significantly (p
- Published
- 2010
16. Principles of membrane protein interactions with annular lipids deduced from aquaporin-0 2D crystals
- Author
-
Thomas Walz, Richard K Hite, and Zongli Li
- Subjects
lens ,Protein Conformation ,Lipid Bilayers ,Aquaporin ,Biology ,010402 general chemistry ,Aquaporins ,Crystallography, X-Ray ,01 natural sciences ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell membrane ,03 medical and health sciences ,chemistry.chemical_compound ,lipid–protein interactions ,Protein structure ,Phosphatidylcholine ,Lens, Crystalline ,medicine ,Escherichia coli ,Animals ,Lipid bilayer ,Eye Proteins ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Crystallography ,Sheep ,General Immunology and Microbiology ,Electron crystallography ,General Neuroscience ,Cell Membrane ,technology, industry, and agriculture ,Membrane Proteins ,Proteins ,Water ,water channel ,Lipids ,0104 chemical sciences ,medicine.anatomical_structure ,electron crystallography ,Biochemistry ,chemistry ,Membrane protein ,Phosphodiester bond ,Biophysics ,lipids (amino acids, peptides, and proteins) ,Dimyristoylphosphatidylcholine - Abstract
We have previously described the interactions of aquaporin-0 (AQP0) with dimyristoyl phosphatidylcholine (DMPC) lipids. We have now determined the 2.5 A structure of AQP0 in two-dimensional (2D) crystals formed with Escherichia coli polar lipids (EPLs), which differ from DMPC both in headgroups and acyl chains. Comparison of the two structures shows that AQP0 does not adapt to the different length of the acyl chains in EPLs and that the distance between the phosphodiester groups in the two leaflets of the DMPC and EPL bilayers is almost identical. The EPL headgroups interact differently with AQP0 than do those of DMPC, but the acyl chains in the EPL and DMPC bilayers occupy similar positions. The interactions of annular lipids with membrane proteins seem to be driven by the propensity of the acyl chains to fill gaps in the protein surface. Interactions of the lipid headgroups may be responsible for the specific interactions found in tightly bound lipids but seem to have a negligible effect on interactions of generic annular lipids with membrane proteins.
- Published
- 2010
17. Structural and functional studies on the stalk of the transferrin receptor
- Author
-
Zongli Li, Danijela Dukovski, Deborah F. Kelly, Eric T. Mack, and Thomas Walz
- Subjects
Models, Molecular ,Protein Conformation ,Iron ,Kinetics ,Biophysics ,Transferrin receptor ,Biochemistry ,Article ,law.invention ,Protein structure ,law ,Receptors, Transferrin ,Functional studies ,Receptor ,Molecular Biology ,chemistry.chemical_classification ,Chemistry ,Cryoelectron Microscopy ,Transferrin ,Cell Biology ,Stalk ,Electron microscope - Abstract
Transferrin (Tf) is an iron carrier protein that consists of two lobes, the N- and C-lobes, which can each bind a Fe(3+) ion. Tf binds to its receptor (TfR), which mediates iron delivery to cells through an endocytotic pathway. Receptor binding facilitates iron release from the Tf C-lobe, but impedes iron release from the N-lobe. An atomic model of the Tf-TfR complex based on single particle electron microscopy (EM) indicated that receptor binding is indeed likely to hinder opening of the N-lobe, thus interfering with its iron release. The atomic model also suggested that the TfR stalks could form additional contacts with the Tf N-lobes, thus potentially further slowing down its iron release. Here, we show that the TfR stalks are unlikely to make strong interactions with the Tf N-lobes and that the stalks have no effect on iron release from the N-lobes of receptor-bound Tf.
- Published
- 2009
18. Ligand binding induces a conformational change in ifnar1 that is propagated to its membrane-proximal domain
- Author
-
Yvonne Becker, Jennifer Julia Strunk, Zongli Li, Jacob Piehler, Eva Jaks, Martynas Gavutis, Peter Lamken, Ingo Gregor, Jörg Enderlein, and Thomas Walz
- Subjects
Receptor complex ,Conformational change ,Chemistry ,Protein Conformation ,Temperature ,Interferon-alpha ,Interferon-beta ,Receptor, Interferon alpha-beta ,Ligand (biochemistry) ,Protein–protein interaction ,Crystallography ,Protein Subunits ,Förster resonance energy transfer ,Ectodomain ,Microscopy, Electron, Transmission ,Structural Biology ,Biophysics ,Fluorescence Resonance Energy Transfer ,Receptor ,Molecular Biology ,Binding domain ,Protein Binding - Abstract
The type I interferon (IFN) receptor plays a key role in innate immunity against viral and bacterial infections. Here, we show by intramolecular Förster resonance energy transfer spectroscopy that ligand binding induces substantial conformational changes in the ectodomain of ifnar1 (ifnar1-EC). Binding of IFN alpha 2 and IFN beta induce very similar conformations of ifnar1, which were confirmed by single-particle electron microscopy analysis of the ternary complexes formed by IFN alpha 2 or IFN beta with the two receptor subunits ifnar1-EC and ifnar2-EC. Photo-induced electron-transfer-based fluorescence quenching and single-molecule fluorescence lifetime measurements revealed that the ligand-induced conformational change in the membrane-distal domains of ifnar1-EC is propagated to its membrane-proximal domain, which is not involved in ligand recognition but is essential for signal activation. Temperature-dependent ligand binding studies as well as stopped-flow fluorescence experiments corroborated a multistep conformational change in ifnar1 upon ligand binding. Our results thus suggest that the relatively intricate architecture of the type I IFN receptor complex is designed to propagate the ligand binding event to and possibly even across the membrane by conformational changes.
- Published
- 2007
19. The EM structure of a type I interferon-receptor complex reveals a novel mechanism for cytokine signaling
- Author
-
Peter Lamken, Thomas Walz, Jacob Piehler, Jennifer Julia Strunk, and Zongli Li
- Subjects
Genetics ,Janus kinase 1 ,Interleukin-17 receptor ,Receptor, Interferon alpha-beta ,Biology ,Glycoprotein 130 ,Article ,Cell biology ,Microscopy, Electron, Transmission ,Structural Biology ,Interleukin-4 receptor ,Interleukin-21 receptor ,Interferon Type I ,Mutation ,Cytokine receptor ,Molecular Biology ,SOCS2 ,Common gamma chain ,Receptors, Interferon ,Signal Transduction - Abstract
Type I interferons have pleiotropic effects, including antiviral, antiproliferative and immunomodulatory responses. All type I interferons bind to a shared receptor consisting of the two transmembrane proteins ifnar1 and ifnar2. We used negative stain electron microscopy to calculate a three-dimensional reconstruction of the ternary complex formed by a triple mutant interferon-α2 with the ectodomains of ifnar1 and ifnar2. We present a model of the complex obtained by placing atomic models of the subunits into the density map of the complex. The complex of interferon-α2 with its receptor, a class II cytokine receptor, shows structural similarities to the complexes formed by growth hormone and erythropoietin with their receptors, members of the class I cytokine receptor family. Despite different assembly mechanisms, class I and class II cytokine receptors thus appear to initiate signaling through similar arrangements of the receptors induced by the binding of their respective ligands.
- Published
- 2007
20. The Hexamer Structure of the Rift Valley Fever Virus Nucleoprotein Suggests a Mechanism for its Assembly into Ribonucleoprotein Complexes
- Author
-
Julien Lescar, Bruno Canard, François Ferron, Bruno Coutard, Dahai Luo, Yeehwa Wong, Rémi N. Charrel, Violaine Lantez, Eric I. Danek, Thomas Walz, and Zongli Li
- Subjects
Models, Molecular ,Macromolecular Assemblies ,Protein Folding ,Viral Diseases ,RNA-binding protein ,Crystallography, X-Ray ,medicine.disease_cause ,Protein structure ,Transcription (biology) ,Macromolecular Structure Analysis ,Biology (General) ,Biomacromolecule-Ligand Interactions ,Macromolecular Complex Analysis ,Polymerase ,Ribonucleoprotein ,0303 health sciences ,biology ,Nucleocapsid Proteins ,Non-coding RNA ,3. Good health ,Cell biology ,Infectious Diseases ,Ribonucleoproteins ,RNA, Viral ,Medicine ,Research Article ,Protein Structure ,DNA, Complementary ,QH301-705.5 ,Viral protein ,Recombinant Fusion Proteins ,Immunology ,Biophysics ,Microbiology ,03 medical and health sciences ,Virology ,Genetics ,medicine ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Biology ,Molecular Biology ,030304 developmental biology ,030306 microbiology ,Virus Assembly ,Computational Biology ,RNA ,RC581-607 ,Surface Plasmon Resonance ,Rift Valley fever virus ,Molecular biology ,Microscopy, Electron ,Mutagenesis, Site-Directed ,biology.protein ,Parasitology ,Immunologic diseases. Allergy ,Protein Multimerization ,Sequence Alignment - Abstract
Rift Valley fever virus (RVFV), a Phlebovirus with a genome consisting of three single-stranded RNA segments, is spread by infected mosquitoes and causes large viral outbreaks in Africa. RVFV encodes a nucleoprotein (N) that encapsidates the viral RNA. The N protein is the major component of the ribonucleoprotein complex and is also required for genomic RNA replication and transcription by the viral polymerase. Here we present the 1.6 Å crystal structure of the RVFV N protein in hexameric form. The ring-shaped hexamers form a functional RNA binding site, as assessed by mutagenesis experiments. Electron microscopy (EM) demonstrates that N in complex with RNA also forms rings in solution, and a single-particle EM reconstruction of a hexameric N-RNA complex is consistent with the crystallographic N hexamers. The ring-like organization of the hexamers in the crystal is stabilized by circular interactions of the N terminus of RVFV N, which forms an extended arm that binds to a hydrophobic pocket in the core domain of an adjacent subunit. The conformation of the N-terminal arm differs from that seen in a previous crystal structure of RVFV, in which it was bound to the hydrophobic pocket in its own core domain. The switch from an intra- to an inter-molecular interaction mode of the N-terminal arm may be a general principle that underlies multimerization and RNA encapsidation by N proteins from Bunyaviridae. Furthermore, slight structural adjustments of the N-terminal arm would allow RVFV N to form smaller or larger ring-shaped oligomers and potentially even a multimer with a super-helical subunit arrangement. Thus, the interaction mode between subunits seen in the crystal structure would allow the formation of filamentous ribonucleocapsids in vivo. Both the RNA binding cleft and the multimerization site of the N protein are promising targets for the development of antiviral drugs., Author Summary The Rift Valley fever virus (RVFV), a negative strand RNA virus spread by infected mosquitoes, affects livestock and humans who can develop a severe disease. We studied the structure of its nucleoprotein (N), which forms a filamentous coat that protects the viral RNA genome and is also required for RNA replication and transcription by the polymerase of the virus. We report the structure of the RVFV N protein at 1.6 Å resolution, which reveals hexameric rings with an external diameter of 100 Å that are formed by exchanges of N-terminal arms between the nearest neighbors. Electron microscopy of recombinant protein in complex with RNA shows that N also forms rings in solution. A reconstruction of the hexameric ring at 25 Å resolution is consistent with the hexamer structure determined by crystallography. We propose that slight structural variations would suffice to convert a ring-shaped oligomer into subunits with a super-helical arrangement and that this mode of protein-protein association forms the basis for the formation of filamentous ribonucleocapsids by this virus family. Both the RNA binding cleft and the multimerization site of the N protein can be targeted for the development of drugs against RVFV.
- Published
- 2011
- Full Text
- View/download PDF
21. Structure of the L Protein of Vesicular Stomatitis Virus from Electron Cryomicroscopy
- Author
-
Nikolaus Grigorieff, Bo Liang, Amal A. Rahmeh, Simon Jenni, Tim Grant, Sean P. J. Whelan, Zongli Li, Stephen C. Harrison, and Benjamin Morin
- Subjects
Models, Molecular ,Transcription, Genetic ,viruses ,Vesicular stomatitis Indiana virus ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Viral Proteins ,chemistry.chemical_compound ,Transcription (biology) ,RNA polymerase ,Polymerase ,Ribonucleoprotein ,Sequence Homology, Amino Acid ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Cryoelectron Microscopy ,RNA ,DNA-Directed RNA Polymerases ,biology.organism_classification ,Molecular biology ,Protein Structure, Tertiary ,RNA silencing ,chemistry ,Biochemistry ,Vesicular stomatitis virus ,biology.protein - Abstract
SummaryThe large (L) proteins of non-segmented, negative-strand RNA viruses, a group that includes Ebola and rabies viruses, catalyze RNA-dependent RNA polymerization with viral ribonucleoprotein as template, a non-canonical sequence of capping and methylation reactions, and polyadenylation of viral messages. We have determined by electron cryomicroscopy the structure of the vesicular stomatitis virus (VSV) L protein. The density map, at a resolution of 3.8 Å, has led to an atomic model for nearly all of the 2109-residue polypeptide chain, which comprises three enzymatic domains (RNA-dependent RNA polymerase [RdRp], polyribonucleotidyl transferase [PRNTase], and methyltransferase) and two structural domains. The RdRp resembles the corresponding enzymatic regions of dsRNA virus polymerases and influenza virus polymerase. A loop from the PRNTase (capping) domain projects into the catalytic site of the RdRp, where it appears to have the role of a priming loop and to couple product elongation to large-scale conformational changes in L.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.