Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Annu Rev Cancer Biol ; 8: 97-113, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38882927

ABSTRACT

Mutations in RAS proteins play a pivotal role in the development of human cancers, driving persistent RAF activation and deregulating the Mitogen-Activated Protein Kinase (MAPK) signaling pathway. While progress has been made in targeting specific oncogenic RAS proteins, effective drug-based therapies for the majority of RAS mutations remain limited. Recent investigations on RAS-RAF complexes and the SHOC2-MRAS-PP1C holoenzyme complex have provided crucial insights into the structural and functional aspects of RAF activation within the MAPK signaling pathway. Moreover, these studies have also unveiled new blueprints for developing inhibitors allowing us to think beyond the current RAS and MEK inhibitors. In this review, we explore the roles of RAS and SHOC2 in activating RAF and discuss potential therapeutic strategies to target these proteins. A comprehensive understanding of the molecular interactions involved in RAF activation and their therapeutic implications holds the potential to drive innovative approaches in combating RAS/RAF-driven cancers.

3.
J Biol Chem ; 299(6): 104789, 2023 06.
Article in English | MEDLINE | ID: mdl-37149146

ABSTRACT

Sprouty-related EVH-1 domain-containing (SPRED) proteins are a family of proteins that negatively regulate the RAS-Mitogen-Activated Protein Kinase (MAPK) pathway, which is involved in the regulation of the mitogenic response and cell proliferation. However, the mechanism by which these proteins affect RAS-MAPK signaling has not been elucidated. Patients with mutations in SPRED give rise to unique disease phenotypes; thus, we hypothesized that distinct interactions across SPRED proteins may account for alternative nodes of regulation. To characterize the SPRED interactome and evaluate how members of the SPRED family function through unique binding partners, we performed affinity purification mass spectrometry. We identified 90-kDa ribosomal S6 kinase 2 (RSK2) as a specific interactor of SPRED2 but not SPRED1 or SPRED3. We identified that the N-terminal kinase domain of RSK2 mediates the interaction between amino acids 123 to 201 of SPRED2. Using X-ray crystallography, we determined the structure of the SPRED2-RSK2 complex and identified the SPRED2 motif, F145A, as critical for interaction. We found that the formation of this interaction is regulated by MAPK signaling events. We also find that this interaction between SPRED2 and RSK2 has functional consequences, whereby the knockdown of SPRED2 resulted in increased phosphorylation of RSK substrates, YB1 and CREB. Furthermore, SPRED2 knockdown hindered phospho-RSK membrane and nuclear subcellular localization. We report that disruption of the SPRED2-RSK complex has effects on RAS-MAPK signaling dynamics. Our analysis reveals that members of the SPRED family have unique protein binding partners and describes the molecular and functional determinants of SPRED2-RSK2 complex dynamics.


Subject(s)
Mitogen-Activated Protein Kinases , Repressor Proteins , Ribosomal Protein S6 Kinases, 90-kDa , Signal Transduction , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/genetics , Humans , Cell Line , Protein Domains , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Gene Knockdown Techniques , Protein Transport/genetics , Protein Binding , Protein Structure, Tertiary , Models, Molecular , Neurofibromin 1/metabolism
4.
Nat Commun ; 14(1): 2275, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37080973

ABSTRACT

Life-threatening bacterial infections in women after childbirth, known as puerperal sepsis, resulted in classical epidemics and remain a global health problem. While outbreaks of puerperal sepsis have been ascribed to Streptococcus pyogenes, little is known about disease mechanisms. Here, we show that the bacterial R28 protein, which is epidemiologically associated with outbreaks of puerperal sepsis, specifically targets the human receptor CEACAM1. This interaction triggers events that would favor the development of puerperal sepsis, including adhesion to cervical cells, suppression of epithelial wound repair and subversion of innate immune responses. High-resolution structural analysis showed that an R28 domain with IgI3-like fold binds to the N-terminal domain of CEACAM1. Together, these findings demonstrate that a single adhesin-receptor interaction can drive the pathogenesis of bacterial sepsis and provide molecular insights into the pathogenesis of one of the most important infectious diseases in medical history.


Subject(s)
Puerperal Infection , Sepsis , Streptococcal Infections , Female , Humans , Pregnancy , Adhesins, Bacterial/genetics , Bacterial Proteins/genetics , Puerperal Infection/epidemiology , Puerperal Infection/microbiology , Sepsis/microbiology , Streptococcal Infections/microbiology , Streptococcus pyogenes
5.
FEBS J ; 290(20): 4852-4863, 2023 10.
Article in English | MEDLINE | ID: mdl-37074066

ABSTRACT

RAF activation is a key step for signalling through the mitogen-activated protein kinase (MAPK) pathway. The SHOC2 protein, along with MRAS and PP1C, forms a high affinity, heterotrimeric holoenzyme that activates RAF kinases by dephosphorylating a specific phosphoserine. Recently, our research, along with that of three other teams, has uncovered valuable structural and functional insights into the SHOC2-MRAS-PP1C (SMP) holoenzyme complex. In this structural snapshot, we review SMP complex assembly, the dependency on the bound-nucleotide state of MRAS, the substitution of MRAS by the canonical RAS proteins and the roles of SHOC2 and MRAS on PP1C activity and specificity. Furthermore, we discuss the effect of several RASopathy mutations identified within the SMP complex and explore potential therapeutic approaches for targeting the SMP complex in RAS/RAF-driven cancers and RASopathies.


Subject(s)
MAP Kinase Signaling System , Mitogen-Activated Protein Kinases , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , ras Proteins/genetics , ras Proteins/metabolism , Holoenzymes/metabolism
6.
Nat Struct Mol Biol ; 29(10): 966-977, 2022 10.
Article in English | MEDLINE | ID: mdl-36175670

ABSTRACT

SHOC2 acts as a strong synthetic lethal interactor with MEK inhibitors in multiple KRAS cancer cell lines. SHOC2 forms a heterotrimeric complex with MRAS and PP1C that is essential for regulating RAF and MAPK-pathway activation by dephosphorylating a specific phosphoserine on RAF kinases. Here we present the high-resolution crystal structure of the SHOC2-MRAS-PP1C (SMP) complex and apo-SHOC2. Our structures reveal that SHOC2, MRAS, and PP1C form a stable ternary complex in which all three proteins synergistically interact with each other. Our results show that dephosphorylation of RAF substrates by PP1C is enhanced upon interacting with SHOC2 and MRAS. The SMP complex forms only when MRAS is in an active state and is dependent on SHOC2 functioning as a scaffolding protein in the complex by bringing PP1C and MRAS together. Our results provide structural insights into the role of the SMP complex in RAF activation and how mutations found in Noonan syndrome enhance complex formation, and reveal new avenues for therapeutic interventions.


Subject(s)
Noonan Syndrome , Humans , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Noonan Syndrome/genetics , Noonan Syndrome/metabolism , Phosphoserine/metabolism , Protein Phosphatase 1 , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/metabolism
7.
Commun Biol ; 4(1): 360, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33742094

ABSTRACT

Human (h) carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) function depends upon IgV-mediated homodimerization or heterodimerization with host ligands, including hCEACAM5, hTIM-3, PD-1, and a variety of microbial pathogens. However, there is little structural information available on how hCEACAM1 transitions between monomeric and dimeric states which in the latter case is critical for initiating hCEACAM1 activities. We therefore mutated residues within the hCEACAM1 IgV GFCC' face including V39, I91, N97, and E99 and examined hCEACAM1 IgV monomer-homodimer exchange using differential scanning fluorimetry, multi-angle light scattering, X-ray crystallography and/or nuclear magnetic resonance. From these studies, we describe hCEACAM1 homodimeric, monomeric and transition states at atomic resolution and its conformational behavior in solution through NMR assignment of the wildtype (WT) hCEACAM1 IgV dimer and N97A mutant monomer. These studies reveal the flexibility of the GFCC' face and its important role in governing the formation of hCEACAM1 dimers and selective heterodimers.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Antigens, CD/chemistry , Antigens, CD/genetics , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Crystallography, X-Ray , Dynamic Light Scattering , Fluorometry , Humans , Magnetic Resonance Spectroscopy , Mutation , Protein Conformation , Protein Multimerization , Structure-Activity Relationship
8.
EMBO J ; 40(7): e106103, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33522633

ABSTRACT

Streptococcus agalactiae, also known as group B Streptococcus (GBS), is the major cause of neonatal sepsis in humans. A critical step to infection is adhesion of bacteria to epithelial surfaces. GBS adhesins have been identified to bind extracellular matrix components and cellular receptors. However, several putative adhesins have no host binding partner characterised. We report here that surface-expressed ß protein of GBS binds to human CEACAM1 and CEACAM5 receptors. A crystal structure of the complex showed that an IgSF domain in ß represents a novel Ig-fold subtype called IgI3, in which unique features allow binding to CEACAM1. Bioinformatic assessment revealed that this newly identified IgI3 fold is not exclusively present in GBS but is predicted to be present in adhesins from other clinically important human pathogens. In agreement with this prediction, we found that CEACAM1 binds to an IgI3 domain found in an adhesin from a different streptococcal species. Overall, our results indicate that the IgI3 fold could provide a broadly applied mechanism for bacteria to target CEACAMs.


Subject(s)
Adhesins, Bacterial/chemistry , Antigens, CD/chemistry , Carcinoembryonic Antigen/chemistry , Cell Adhesion Molecules/chemistry , Adhesins, Bacterial/metabolism , Animals , Antigens, CD/metabolism , Binding Sites , CHO Cells , Carcinoembryonic Antigen/metabolism , Cell Adhesion Molecules/metabolism , Cricetinae , Cricetulus , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/metabolism , HeLa Cells , Humans , Protein Binding , Streptococcus agalactiae/metabolism
9.
Adv Exp Med Biol ; 1149: 57-75, 2019.
Article in English | MEDLINE | ID: mdl-31016628

ABSTRACT

Helicobacter pylori adherence to host epithelial cells is essential for its survival against the harsh conditions of the stomach and for successful colonization. Adherence of H. pylori is achieved through several related families of outer membrane proteins and proteins of a type IV secretion system (T4SS), which bridge H. pylori to host cells through protein-protein and other protein-ligand interactions. Local environmental conditions such as cell type, available host cell surface proteins and/or ligands, as well as responses by the host immune system force H. pylori to alter expression of these proteins to adapt quickly to the local environment in order to colonize and survive. Some of these host-pathogen interactions appear to function in a "catch-and-release" manner, regulated by reversible binding at varying pH and allowing H. pylori to detach itself from cells or debris sloughed off the gastric epithelial lining in order to return for subsequent productive interactions. Other interactions between bacterial adhesin proteins and host adhesion molecules, however, appear to function as a committed step in certain pathogenic processes, such as translocation of the CagA oncoprotein through the H. pylori T4SS and into host gastric epithelial cells. Understanding these adhesion interactions is critical for devising new therapeutic strategies, as they are responsible for the earliest stage of infection and its maintenance. This review will discuss the expression and regulation of several outer membrane proteins and CagL, how they engage their known host cell protein/ligand targets, and their effects on clinical outcome.


Subject(s)
Bacterial Adhesion , Epithelial Cells , Helicobacter Infections , Helicobacter pylori , Host-Pathogen Interactions , Stomach , Adhesins, Bacterial/metabolism , Epithelial Cells/microbiology , Humans , Stomach/microbiology
10.
Sci Rep ; 8(1): 17512, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504845

ABSTRACT

T-cell immunoglobulin and mucin domain containing protein-3 (TIM-3) is an important immune regulator. Here, we describe a novel high resolution (1.7 Å) crystal structure of the human (h)TIM-3 N-terminal variable immunoglobulin (IgV) domain with bound calcium (Ca++) that was confirmed by nuclear magnetic resonance (NMR) spectroscopy. Significant conformational differences were observed in the B-C, C'-C″ and C'-D loops of hTIM-3 compared to mouse (m)TIM-3, hTIM-1 and hTIM-4. Further, the conformation of the C-C' loop of hTIM-3 was notably different from hTIM-4. Consistent with the known metal ion-dependent binding of phosphatidylserine (PtdSer) to mTIM-3 and mTIM-4, the NMR spectral analysis and crystal structure of Ca++-bound hTIM-3 revealed that residues in the hTIM-3 F-G loop coordinate binding to Ca++. In addition, we established a novel biochemical assay to define hTIM-3 functionality as determined by binding to human carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1). These studies provide new insights useful for understanding and targeting hTIM-3.


Subject(s)
Crystallography, X-Ray/methods , Hepatitis A Virus Cellular Receptor 2/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Humans , Mice , Protein Conformation , Sequence Homology, Amino Acid
11.
EMBO J ; 37(13)2018 07 02.
Article in English | MEDLINE | ID: mdl-29724755

ABSTRACT

Helicobacter pylori infects half of the world's population, and strains that encode the cag type IV secretion system for injection of the oncoprotein CagA into host gastric epithelial cells are associated with elevated levels of cancer. CagA translocation into host cells is dependent on interactions between the H. pylori adhesin protein HopQ and human CEACAMs. Here, we present high-resolution structures of several HopQ-CEACAM complexes and CEACAMs in their monomeric and dimeric forms establishing that HopQ uses a coupled folding and binding mechanism to engage the canonical CEACAM dimerization interface for CEACAM recognition. By combining mutagenesis with biophysical and functional analyses, we show that the modes of CEACAM recognition by HopQ and CEACAMs themselves are starkly different. Our data describe precise molecular mechanisms by which microbes exploit host CEACAMs for infection and enable future development of novel oncoprotein translocation inhibitors and H. pylori-specific antimicrobial agents.


Subject(s)
Antigens, Bacterial/physiology , Antigens, CD/physiology , Bacterial Proteins/physiology , Cell Adhesion Molecules/physiology , Helicobacter pylori/physiology , Oncogene Proteins/physiology , Antigens, CD/chemistry , Bacterial Proteins/chemistry , Cell Adhesion Molecules/chemistry , HEK293 Cells , Humans , Mutagenesis , Protein Multimerization , Protein Transport
12.
Immunity ; 47(3): 510-523.e4, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28930661

ABSTRACT

Within the interleukin 1 (IL-1) cytokine family, IL-1 receptor accessory protein (IL-1RAcP) is the co-receptor for eight receptor-cytokine pairs, including those involving cytokines IL-1ß and IL-33. Unlike IL-1ß, IL-33 does not have a signaling complex that includes both its cognate receptor, ST2, and the shared co-receptor IL-1RAcP, which we now present here. Although the IL-1ß and IL-33 complexes shared structural features and engaged identical molecular surfaces of IL-1RAcP, these cytokines had starkly different strategies for co-receptor engagement and signal activation. Our data suggest that IL-1ß binds to IL-1RI to properly present the cytokine to IL-1RAcP, whereas IL-33 binds to ST2 in order to conformationally constrain the cognate receptor in an IL-1RAcP-receptive state. These findings indicate that members of the IL-1 family of cytokines use distinct molecular mechanisms to signal through their shared co-receptor, and they provide the foundation from which to design new therapies to target IL-33 signaling.


Subject(s)
Interleukin-1/metabolism , Receptors, Interleukin-1/metabolism , Signal Transduction , Animals , Binding Sites , Humans , Hydrophobic and Hydrophilic Interactions , Interleukin-1/chemistry , Interleukin-1 Receptor-Like 1 Protein/chemistry , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/chemistry , Interleukin-33/metabolism , Mice , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutation , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/genetics
13.
Methods Mol Biol ; 1535: 77-93, 2017.
Article in English | MEDLINE | ID: mdl-27914074

ABSTRACT

Employing biophysical and structural methods is a powerful way to elucidate mechanisms of molecular recognition in bacterial pathogenesis. Such studies invariably depend on the production of pure, folded and stable proteins. Many proteins that can be expressed recombinantly ultimately fail to meet one or more of these criteria. The cag proteins from Helicobacter pylori form a secretion system that delivers the oncoprotein, CagA, into human gastric epithelial cells through an interaction between CagL and host cell integrins, where it can cause gastric adenocarcinoma. Expression of full length CagA and CagL is problematic as CagA undergoes rapid degradation during purification and CagL is thermally unstable. Here, we describe a method for the purification of CagA that results in the production of the full length protein through coexpression with its endogenous chaperone, CagF, and its subsequent separation from its chaperone. Furthermore, we detail the production of CagL and the use of differential scanning fluorimetry to identify how CagL is thermally stabilized by reduced pH, which led to a new crystal form of CagL and novel insight to pathogenic mechanisms. The methods described here for the production of stable cag proteins can be applied to a wide range of proteins involved in bacterial pathogenesis.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Helicobacter pylori/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Cloning, Molecular , Gene Expression , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Humans , Plasmids/genetics , Protein Stability , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Spectrometry, Fluorescence , Structure-Activity Relationship
14.
Sci Rep ; 6: 36685, 2016 11 09.
Article in English | MEDLINE | ID: mdl-27827447

ABSTRACT

Antibody-dependent cell-mediated cytotoxicity (ADCC) by non-neutralizing antibodies (nnAbs) specific to the HIV envelope (Env) glycoproteins present at the surface of virus sensitized or infected cells plays a role in the effective adaptive immune response to HIV. Here, we explore the molecular basis for the epitope at the disulfide loop region (DLR) of the principal immunodominant domain of gp41, recognized by the well-known nnAb F240. Our structural studies reveal details of the F240-gp41 interface and describe a structure of DLR that is distinct from known conformations of this region studied in the context of either CD4-unliganded Env trimer or the gp41 peptide in the unbound state. These data coupled with binding and functional analyses indicate that F240 recognizes non-trimeric Env forms which are significantly overexpressed on intact virions but poorly represented at surfaces of cells infected with infectious molecular clones and endogenously-infected CD4 T cells from HIV-1-infected individuals. Furthermore, although we detect ADCC activities of F240 against cells spinoculated with intact virions, our data suggest that these activities result from F240 recognition of gp41 stumps or misfolded Env variants present on virions rather than its ability to recognize functional gp41 transition structures emerging on trimeric Env post CD4 receptor engagement.


Subject(s)
Epitopes/chemistry , HIV Antibodies/chemistry , HIV Envelope Protein gp41/chemistry , HIV-1/chemistry , Single-Chain Antibodies/chemistry , Surface Plasmon Resonance , Binding Sites, Antibody , Epitopes/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp41/immunology , HIV-1/immunology , Humans , Peptides/chemistry , Peptides/immunology , Single-Chain Antibodies/immunology
16.
Nat Microbiol ; 2: 16188, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27748756

ABSTRACT

Helicobacter pylori (Hp) strains that carry the cag type IV secretion system (cag-T4SS) to inject the cytotoxin-associated antigen A (CagA) into host cells are associated with peptic ulcer disease and gastric adenocarcinoma. CagA translocation by Hp is mediated by ß1 integrin interaction of the cag-T4SS. However, other cellular receptors or bacterial outer membrane adhesins essential for this process are unknown. Here, we identify the HopQ protein as a genuine Hp adhesin, exploiting defined members of the carcinoembryonic antigen-related cell adhesion molecule family (CEACAMs) as host cell receptors. HopQ binds the amino-terminal IgV-like domain of human CEACAM1, CEACAM3, CEACAM5 or CEACAM6 proteins, thereby enabling translocation of the major pathogenicity factor CagA into host cells. The HopQ-CEACAM interaction is characterized by a remarkably high affinity (KD from 23 to 268 nM), which is independent of CEACAM glycosylation, identifying CEACAMs as bona fide protein receptors for Hp. Our data suggest that the HopQ-CEACAM interaction contributes to gastric colonization or Hp-induced pathologies, although the precise role and functional consequences of this interaction in vivo remain to be determined.


Subject(s)
Adhesins, Bacterial/metabolism , Antigens, Bacterial/metabolism , Bacterial Adhesion , Bacterial Proteins/metabolism , Cell Adhesion Molecules/metabolism , Helicobacter pylori/physiology , Host-Pathogen Interactions , Protein Transport , Cell Line , Humans , Protein Binding
17.
Elife ; 52016 09 24.
Article in English | MEDLINE | ID: mdl-27664419

ABSTRACT

Flagella are crucial for bacterial motility and pathogenesis. The flagellar capping protein (FliD) regulates filament assembly by chaperoning and sorting flagellin (FliC) proteins after they traverse the hollow filament and exit the growing flagellum tip. In the absence of FliD, flagella are not formed, resulting in impaired motility and infectivity. Here, we report the 2.2 Å resolution X-ray crystal structure of FliD from Pseudomonas aeruginosa, the first high-resolution structure of any FliD protein from any bacterium. Using this evidence in combination with a multitude of biophysical and functional analyses, we find that Pseudomonas FliD exhibits unexpected structural similarity to other flagellar proteins at the domain level, adopts a unique hexameric oligomeric state, and depends on flexible determinants for oligomerization. Considering that the flagellin filaments on which FliD oligomers are affixed vary in protofilament number between bacteria, our results suggest that FliD oligomer stoichiometries vary across bacteria to complement their filament assemblies.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Protein Multimerization , Pseudomonas aeruginosa/enzymology , Crystallography, X-Ray , Flagellin/metabolism , Models, Molecular , Protein Conformation
18.
Proc Natl Acad Sci U S A ; 112(44): 13561-6, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26483485

ABSTRACT

Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) comprise a large family of cell surface adhesion molecules that bind to themselves and other family members to carry out numerous cellular functions, including proliferation, signaling, differentiation, tumor suppression, and survival. They also play diverse and significant roles in immunity and infection. The formation of CEACAM oligomers is caused predominantly by interactions between their N-terminal IgV domains. Although X-ray crystal structures of CEACAM IgV domain homodimers have been described, how CEACAMs form heterodimers or remain monomers is poorly understood. To address this key aspect of CEACAM function, we determined the crystal structures of IgV domains that form a homodimeric CEACAM6 complex, monomeric CEACAM8, and a heterodimeric CEACAM6-CEACAM8 complex. To confirm and quantify these interactions in solution, we used analytical ultracentrifugation to measure the dimerization constants of CEACAM homodimers and isothermal titration calorimetry to determine the thermodynamic parameters and binding affinities of CEACAM heterodimers. We found the CEACAM6-CEACAM8 heterodimeric state to be substantially favored energetically relative to the CEACAM6 homodimer. Our data provide a molecular basis for the adoption of the diverse oligomeric states known to exist for CEACAMs and suggest ways in which CEACAM6 and CEACAM8 regulate the biological functions of one another, as well as of additional CEACAMs with which they interact, both in cis and in trans.


Subject(s)
Antigens, CD/chemistry , Cell Adhesion Molecules/chemistry , Protein Multimerization , Protein Structure, Tertiary , Amino Acid Sequence , Antigens, CD/genetics , Antigens, CD/metabolism , Calorimetry/methods , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Crystallography, X-Ray , Epithelial Cells/metabolism , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Granulocytes/metabolism , HEK293 Cells , Humans , Hydrogen Bonding , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Binding , Sequence Homology, Amino Acid
19.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 9): 1169-75, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26323304

ABSTRACT

CEACAM7 is a human cellular adhesion protein that is expressed on the surface of colon and rectum epithelial cells and is downregulated in colorectal cancers. It achieves cell adhesion through dimerization of the N-terminal IgV domain. The crystal structure of the N-terminal dimerization domain of CEACAM has been determined at 1.47 Šresolution. The overall fold of CEACAM7 is similar to those of CEACAM1 and CEACAM5; however, there are differences, the most notable of which is an insertion that causes the C'' strand to buckle, leading to the creation of a hydrogen bond in the dimerization interface. The Kdimerization for CEACAM7 determined by sedimentation equilibrium is tenfold tighter than that measured for CEACAM5. These findings suggest that the dimerization affinities of CEACAMs are modulated via sequence variation in the dimerization surface.


Subject(s)
Carcinoembryonic Antigen/chemistry , Protein Multimerization , Amino Acid Sequence , GPI-Linked Proteins/chemistry , Humans , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Tertiary , Sequence Alignment
20.
Biochemistry ; 54(21): 3337-47, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25963096

ABSTRACT

The leading risk factor for gastric cancer in humans is infection by Helicobacter pylori strains that express and translocate the oncoprotein CagA into host epithelial cells. Once inside host cells, CagA interacts with ASPP2, which specifically stimulates p53-mediated apoptosis and reverses its pro-apoptotic function to promote ASPP2-dependent degradation of p53. The X-ray crystal structure of a complex between the N-terminal domain of CagA and a 56-residue fragment of ASPP2, of which 22 residues were resolved, was recently described. Here, we present biochemical and biophysical analyses of the interaction between the additional regions of CagA and ASPP2 potentially involved in this interaction. Using size exclusion chromatography-multiangle laser light scattering, circular dichroism, and nuclear magnetic resonance analyses, we observed that the ASPP2 region spanning residues 331-692, which was not part of the ASPP2 fragment used for crystallization, is intrinsically disordered in its unbound state. By surface plasmon resonance analysis and isothermal titration calorimetry, we found that a portion of this disordered region in ASPP2, residues 448-692, binds to the N-terminal domain of CagA. We also measured the affinity of the complex between the ASPP2 fragment composed of residues 693-918 and inclusive of the fragment used for crystallization and CagA. Additionally, we mapped the binding regions between ASPP2 and CagA using peptide arrays, demonstrating interactions between CagA and numerous peptides distributed throughout the ASPP2 protein sequence. Our results identify previously uncharacterized regions distributed throughout the protein sequence of ASPP2 as determinants of CagA binding, providing mechanistic insight into apoptosis reprogramming by CagA and potential new drug targets for H. pylori-mediated gastric cancer.


Subject(s)
Antigens, Bacterial/metabolism , Apoptosis Regulatory Proteins/metabolism , Bacterial Proteins/metabolism , Helicobacter Infections/complications , Helicobacter pylori/metabolism , Stomach Neoplasms/microbiology , Antigens, Bacterial/chemistry , Apoptosis Regulatory Proteins/chemistry , Bacterial Proteins/chemistry , Humans , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Stomach Neoplasms/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...