Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38645081

ABSTRACT

The estrogen receptor-α (ER) is thought to function only as a homodimer, but responds to a variety of environmental, metazoan, and therapeutic estrogens at sub-saturating doses, supporting binding mixtures of ligands as well as dimers that are only partially occupied. Here, we present a series of flexible ER ligands that bind to receptor dimers with individual ligand poses favoring distinct receptor conformations -receptor conformational heterodimers-mimicking the binding of two different ligands. Molecular dynamics simulations showed that the pairs of different ligand poses changed the correlated motion across the dimer interface to generate asymmetric communication between the dimer interface, the ligands, and the surface binding sites for epigenetic regulatory proteins. By examining binding of the same ligand in crystal structures of ER in the agonist versus antagonist conformers, we also showed that these allosteric signals are bidirectional. The receptor conformer can drive different ligand binding modes to support agonist versus antagonist activity profiles, a revision of ligand binding theory that has focused on unidirectional signaling from ligand to the coregulator binding site. We also observed differences in the allosteric signals between ligand and coregulator binding sites in the monomeric versus dimeric receptor, and when bound by two different ligands, states that are physiologically relevant. Thus, ER conformational heterodimers integrate two different ligand-regulated activity profiles, representing new modes for ligand-dependent regulation of ER activity. Significance: The estrogen receptor-α (ER) regulates transcription in response to a hormonal milieu that includes low levels of estradiol, a variety of environmental estrogens, as well as ER antagonists such as breast cancer anti-hormonal therapies. While ER has been studied as a homodimer, the variety of ligand and receptor concentrations in different tissues means that the receptor can be occupied with two different ligands, with only one ligand in the dimer, or as a monomer. Here, we use X-ray crystallography and molecular dynamics simulations to reveal a new mode for ligand regulation of ER activity whereby sequence-identical homodimers can act as functional or conformational heterodimers having unique signaling characteristics, with ligand-selective allostery operating across the dimer interface integrating two different signaling outcomes.

2.
Int J Mol Sci ; 24(11)2023 May 29.
Article in English | MEDLINE | ID: mdl-37298410

ABSTRACT

Plakophilin-3 is a ubiquitously expressed protein found widely in epithelial cells and is a critical component of desmosomes. The plakophilin-3 carboxy-terminal domain harbors nine armadillo repeat motifs with largely unknown functions. Here, we report the 5 Å cryogenic electron microscopy (cryoEM) structure of the armadillo repeat motif domain of plakophilin-3, one of the smaller cryoEM structures reported to date. We find that this domain is a monomer or homodimer in solution. In addition, using an in vitro actin co-sedimentation assay, we show that the armadillo repeat domain of plakophilin-3 directly interacts with F-actin. This feature, through direct interactions with actin filaments, could be responsible for the observed association of extra-desmosomal plakophilin-3 with the actin cytoskeleton directly attached to the adherens junctions in A431 epithelial cells. Further, we demonstrate, through lipid binding analyses, that plakophilin-3 can effectively be recruited to the plasma membrane through phosphatidylinositol-4,5-bisphosphate-mediated interactions. Collectively, we report on novel properties of plakophilin-3, which may be conserved throughout the plakophilin protein family and may be behind the roles of these proteins in cell-cell adhesion.


Subject(s)
Actins , Plakophilins , Actin Cytoskeleton , Actins/metabolism , Desmosomes/metabolism , Plakophilins/metabolism
3.
Commun Biol ; 6(1): 276, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36928388

ABSTRACT

Attachment between cells is crucial for almost all aspects of the life of cells. These inter-cell adhesions are mediated by the binding of transmembrane cadherin receptors of one cell to cadherins of a neighboring cell. Inside the cell, cadherin binds ß-catenin, which interacts with α-catenin. The transitioning of cells between migration and adhesion is modulated by α-catenin, which links cell junctions and the plasma membrane to the actin cytoskeleton. At cell junctions, a single ß-catenin/α-catenin heterodimer slips along filamentous actin in the direction of cytoskeletal tension which unfolds clustered heterodimers to form catch bonds with F-actin. Outside cell junctions, α-catenin dimerizes and links the plasma membrane to F-actin. Under cytoskeletal tension, α-catenin unfolds and forms an asymmetric catch bond with F-actin. To understand the mechanism of this important α-catenin function, we determined the 2.7 Å cryogenic electron microscopy (cryoEM) structures of filamentous actin alone and bound to human dimeric α-catenin. Our structures provide mechanistic insights into the role of the α-catenin interdomain interactions in directing α-catenin function and suggest a bivalent mechanism. Further, our cryoEM structure of human monomeric α-catenin provides mechanistic insights into α-catenin autoinhibition. Collectively, our structures capture the initial α-catenin interaction with F-actin before the sensing of force, which is a crucial event in cell adhesion and human disease.


Subject(s)
Actin Cytoskeleton , Intercellular Junctions , alpha Catenin , Humans , Actin Cytoskeleton/metabolism , Actins/metabolism , alpha Catenin/chemistry , alpha Catenin/metabolism , beta Catenin/metabolism , Cadherins/metabolism , Intercellular Junctions/metabolism
4.
J Biol Chem ; 299(2): 102817, 2023 02.
Article in English | MEDLINE | ID: mdl-36539037

ABSTRACT

The regulation of cell-cell junctions during epidermal morphogenesis ensures tissue integrity, a process regulated by α-catenin. This cytoskeletal protein connects the cadherin complex to filamentous actin at cell-cell junctions. The cadherin-catenin complex plays key roles in cell physiology, organism development, and disease. While mutagenesis of Caenorhabditis elegans cadherin and catenin shows that these proteins are key for embryonic morphogenesis, we know surprisingly little about their structure and attachment to the cytoskeleton. In contrast to mammalian α-catenin that functions as a dimer or monomer, the α-catenin ortholog from C. elegans, HMP1 for humpback, is a monomer. Our cryogenic electron microscopy (cryoEM) structure of HMP1/α-catenin reveals that the amino- and carboxy-terminal domains of HMP1/α-catenin are disordered and not in contact with the remaining HMP1/α-catenin middle domain. Since the carboxy-terminal HMP1/α-catenin domain is the F-actin-binding domain (FABD), this interdomain constellation suggests that HMP1/α-catenin is constitutively active, which we confirm biochemically. Our perhaps most surprising finding, given the high sequence similarity between the mammalian and nematode proteins, is our cryoEM structure of HMP1/α-catenin bound to F-actin. Unlike the structure of mammalian α-catenin bound to F-actin, binding to F-actin seems to allosterically convert a loop region of the HMP1/α-catenin FABD to extend an HMP1/α-catenin FABD α-helix. We use cryoEM and bundling assays to show for the first time how the FABD of HMP1/α-catenin bundles actin in the absence of force. Collectively, our data advance our understanding of α-catenin regulation of cell-cell contacts and additionally aid our understanding of the evolution of multicellularity in metazoans.


Subject(s)
Actin Cytoskeleton , Caenorhabditis elegans , alpha Catenin , Animals , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Actins/chemistry , Actins/metabolism , Actins/ultrastructure , alpha Catenin/chemistry , alpha Catenin/metabolism , Cadherins/metabolism , Mammals , Protein Conformation, alpha-Helical , Protein Domains , Cryoelectron Microscopy , Cell Adhesion , Cell Communication
5.
Matrix Biol Plus ; 12: 100074, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34337379

ABSTRACT

The Neurofibromatosis type 2 gene encodes the Nf2/merlin tumor suppressor protein that is responsible for the regulation of cell proliferation. Once activated, Nf2/merlin modulates adhesive signaling pathways and thereby inhibits cell growth. Nf2/merlin controls oncogenic gene expression by modulating the Hippo pathway. By responding to several physical and biochemical stimuli, Hippo signaling determines contact inhibition of proliferation as well as organ size. The large tumor suppressor (LATS) serine/threonine-protein kinase is the key enzyme in the highly conserved kinase cascade that negatively regulates the activity and localization of the transcriptional coactivators Yes-associated protein (YAP) and its paralogue transcriptional coactivator with PDZ-binding motif (TAZ). Nf2/merlin belongs to the band 4.1, ezrin, radixin, moesin (FERM) gene family that links the actin cytoskeleton to adherens junctions, remodels adherens junctions during epithelial morphogenesis and maintains organized apical surfaces on the plasma cell membrane. Nf2/merlin and ERM proteins have a globular N-terminal cloverleaf head domain, the FERM domain, that binds to the plasma membrane, a central α-helical domain, and a tail domain that binds to its head domain. Here we present the high-resolution crystal structure of Nf2/merlin bound to LATS1 which shows that LATS1 binding to Nf2/merlin displaces the Nf2/merlin tail domain and causes an allosteric shift in the Nf2/merlin α-helix that extends from its FERM domain. This is consistent with the fact that full-length Nf2/merlin binds LATS1 ~10-fold weaker compared to LATS1 binding to the Nf2/merlin-PIP2 complex. Our data increase our understanding of Nf2/merlin biology by providing mechanistic insights into the Hippo pathway that are relevant to several diseases in particular oncogenic features that are associated with cancers.

6.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34452998

ABSTRACT

Efforts to improve estrogen receptor-α (ER)-targeted therapies in breast cancer have relied upon a single mechanism, with ligands having a single side chain on the ligand core that extends outward to determine antagonism of breast cancer growth. Here, we describe inhibitors with two ER-targeting moieties, one of which uses an alternate structural mechanism to generate full antagonism, freeing the side chain to independently determine other critical properties of the ligands. By combining two molecular targeting approaches into a single ER ligand, we have generated antiestrogens that function through new mechanisms and structural paradigms to achieve antagonism. These dual-mechanism ER inhibitors (DMERIs) cause alternate, noncanonical structural perturbations of the receptor ligand-binding domain (LBD) to antagonize proliferation in ER-positive breast cancer cells and in allele-specific resistance models. Our structural analyses with DMERIs highlight marked differences from current standard-of-care, single-mechanism antiestrogens. These findings uncover an enhanced flexibility of the ER LBD through which it can access nonconsensus conformational modes in response to DMERI binding, broadly and effectively suppressing ER activity.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Crystallography, X-Ray , Female , Humans , Protein Binding , Protein Conformation , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Int J Mol Sci ; 22(2)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33440717

ABSTRACT

Vinculin and its heart-specific splice variant metavinculin are key regulators of cell adhesion processes. These membrane-bound cytoskeletal proteins regulate the cell shape by binding to several other proteins at cell-cell and cell-matrix junctions. Vinculin and metavinculin link integrin adhesion molecules to the filamentous actin network. Loss of both proteins prevents cell adhesion and cell spreading and reduces the formation of stress fibers, focal adhesions, or lamellipodia extensions. The binding of talin at cell-matrix junctions or of α-catenin at cell-cell junctions activates vinculin and metavinculin by releasing their autoinhibitory head-tail interaction. Once activated, vinculin and metavinculin bind F-actin via their five-helix bundle tail domains. Unlike vinculin, metavinculin has a 68-amino-acid insertion before the second α-helix of this five-helix F-actin-binding domain. Here, we present the full-length cryogenic electron microscopy structure of metavinculin that captures the dynamics of its individual domains and unveiled a hallmark structural feature, namely a kinked isoform-specific α-helix in its F-actin-binding domain. Our identified conformational landscape of metavinculin suggests a structural priming mechanism that is consistent with the cell adhesion functions of metavinculin in response to mechanical and cellular cues. Our findings expand our understanding of metavinculin function in the heart with implications for the etiologies of cardiomyopathies.


Subject(s)
Cryoelectron Microscopy , Cytoskeleton/chemistry , Protein Interaction Domains and Motifs , Vinculin/chemistry , Actins/chemistry , Actins/metabolism , Cell Adhesion , Cell Membrane/chemistry , Cell Membrane/metabolism , Crystallography, X-Ray , Cytoskeleton/metabolism , Humans , Models, Molecular , Peptides , Protein Binding , Protein Conformation , Structure-Activity Relationship , Vinculin/metabolism
8.
Nat Commun ; 11(1): 6013, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33243994

ABSTRACT

SARS-CoV-2 variants with spike (S)-protein D614G mutations now predominate globally. We therefore compare the properties of the mutated S protein (SG614) with the original (SD614). We report here pseudoviruses carrying SG614 enter ACE2-expressing cells more efficiently than those with SD614. This increased entry correlates with less S1-domain shedding and higher S-protein incorporation into the virion. Similar results are obtained with virus-like particles produced with SARS-CoV-2 M, N, E, and S proteins. However, D614G does not alter S-protein binding to ACE2 or neutralization sensitivity of pseudoviruses. Thus, D614G may increase infectivity by assembling more functional S protein into the virion.


Subject(s)
COVID-19/virology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/genetics , Virion/metabolism , Virus Assembly/genetics , Virus Internalization , Amino Acid Substitution , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/epidemiology , HEK293 Cells , Humans , Mutation , Pandemics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/metabolism
9.
J Biol Chem ; 295(37): 12885-12899, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32605925

ABSTRACT

Integrin receptors regulate normal cellular processes such as signaling, cell migration, adhesion to the extracellular matrix, and leukocyte function. Talin recruitment to the membrane is necessary for its binding to and activation of integrin. Vertebrates have two highly conserved talin homologs that differ in their expression patterns. The F1-F3 FERM subdomains of cytoskeletal proteins resemble a cloverleaf, but in talin1, its F1 subdomain and additional F0 subdomain align more linearly with its F2 and F3 subdomains. Here, we present the talin2 crystal structure, revealing that its F0-F1 di-subdomain displays another unprecedented constellation, whereby the F0-F1-F2 adopts a new cloverleaf-like arrangement. Using multiangle light scattering (MALS), fluorescence lifetime imaging (FLIM), and FRET analyses, we found that substituting the corresponding residues in talin2 that abolish lipid binding in talin1 disrupt the binding of talin to the membrane, focal adhesion formation, and cell spreading. Our results provide the molecular details of the functions of specific talin isoforms in cell adhesion.


Subject(s)
Cell Adhesion , Focal Adhesions , Talin , Cell Line , Focal Adhesions/chemistry , Focal Adhesions/genetics , Focal Adhesions/metabolism , Humans , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Talin/chemistry , Talin/genetics , Talin/metabolism
10.
bioRxiv ; 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32587973

ABSTRACT

SARS coronavirus 2 (SARS-CoV-2) isolates encoding a D614G mutation in the viral spike (S) protein predominate over time in locales where it is found, implying that this change enhances viral transmission. We therefore compared the functional properties of the S proteins with aspartic acid (S D614 ) and glycine (S G614 ) at residue 614. We observed that retroviruses pseudotyped with S G614 infected ACE2-expressing cells markedly more efficiently than those with S D614 . This greater infectivity was correlated with less S1 shedding and greater incorporation of the S protein into the pseudovirion. Similar results were obtained using the virus-like particles produced with SARS-CoV-2 M, N, E, and S proteins. However, S G614 did not bind ACE2 more efficiently than S D614 , and the pseudoviruses containing these S proteins were neutralized with comparable efficiencies by convalescent plasma. These results show S G614 is more stable than S D614 , consistent with epidemiological data suggesting that viruses with S G614 transmit more efficiently.

12.
Elife ; 72018 12 12.
Article in English | MEDLINE | ID: mdl-30540250

ABSTRACT

Signaling by the G-protein-coupled receptors (GPCRs) plays fundamental role in a vast number of essential physiological functions. Precise control of GPCR signaling requires action of regulators of G protein signaling (RGS) proteins that deactivate heterotrimeric G proteins. RGS proteins are elaborately regulated and comprise multiple domains and subunits, yet structural organization of these assemblies is poorly understood. Here, we report a crystal structure and dynamics analyses of the multisubunit complex of RGS7, a major regulator of neuronal signaling with key roles in controlling a number of drug target GPCRs and links to neuropsychiatric disease, metabolism, and cancer. The crystal structure in combination with molecular dynamics and mass spectrometry analyses reveals unique organizational features of the complex and long-range conformational changes imposed by its constituent subunits during allosteric modulation. Notably, several intermolecular interfaces in the complex work in synergy to provide coordinated modulation of this key GPCR regulator.


Subject(s)
Carrier Proteins/chemistry , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Proteins/metabolism , Molecular Dynamics Simulation , Neurons/metabolism , RGS Proteins/chemistry , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Crystallography, X-Ray , Deuterium Exchange Measurement , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mass Spectrometry , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Conformation , Protein Multimerization , RGS Proteins/genetics , RGS Proteins/metabolism , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera
13.
Proc Natl Acad Sci U S A ; 115(41): 10339-10344, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30254158

ABSTRACT

Multicellular organisms have well-defined, tightly regulated mechanisms for cell adhesion. Heterodimeric αß integrin receptors play central roles in this function and regulate processes for normal cell functions, including signaling, cell migration, and development, binding to the extracellular matrix, and senescence. They are involved in hemostasis and the immune response, participate in leukocyte function, and have biological implications in angiogenesis and cancer. Proper control of integrin activation for cellular communication with the external environment requires several physiological processes. Perturbation of these equilibria may lead to constitutive integrin activation that results in bleeding disorders. Furthermore, integrins play key roles in cancer progression and metastasis in which certain tumor types exhibit higher levels of various integrins. Thus, the integrin-associated signaling complex is important for cancer therapy development. During inside-out signaling, the cytoskeletal protein talin plays a key role in regulating integrin affinity whereby the talin head domain activates integrin by binding to the cytoplasmic tail of ß-integrin and acidic membrane phospholipids. To understand the mechanism of integrin activation by talin, we determined the crystal structure of the talin head domain bound to the acidic phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2), allowing us to design a lipid-binding-deficient talin mutant. Our confocal microscopy with talin knockout cells suggests that the talin-cell membrane interaction seems essential for focal adhesion formation and stabilization. Basal integrin activation in Chinese hamster ovary cells suggests that the lipid-binding-deficient talin mutant inhibits integrin activation. Thus, membrane attachment of talin seems necessary for integrin activation and focal adhesion formation.


Subject(s)
Cell Membrane/metabolism , Focal Adhesions/physiology , Integrins/metabolism , Talin/chemistry , Talin/metabolism , Allosteric Regulation , Animals , Binding Sites , CHO Cells , Cricetulus , Crystallography, X-Ray , Models, Molecular , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Conformation , Protein Domains , Talin/genetics
14.
Immunity ; 47(6): 1182-1196.e10, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29262351

ABSTRACT

CD4+ T cells are tightly regulated by microbiota in the intestine, but whether intestinal T cells interface with host-derived metabolites is less clear. Here, we show that CD4+ T effector (Teff) cells upregulated the xenobiotic transporter, Mdr1, in the ileum to maintain homeostasis in the presence of bile acids. Whereas wild-type Teff cells upregulated Mdr1 in the ileum, those lacking Mdr1 displayed mucosal dysfunction and induced Crohn's disease-like ileitis following transfer into Rag1-/- hosts. Mdr1 mitigated oxidative stress and enforced homeostasis in Teff cells exposed to conjugated bile acids (CBAs), a class of liver-derived emulsifying agents that actively circulate through the ileal mucosa. Blocking ileal CBA reabsorption in transferred Rag1-/- mice restored Mdr1-deficient Teff cell homeostasis and attenuated ileitis. Further, a subset of ileal Crohn's disease patients displayed MDR1 loss of function. Together, these results suggest that coordinated interaction between mucosal Teff cells and CBAs in the ileum regulate intestinal immune homeostasis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology , Bile Acids and Salts/immunology , CD4-Positive T-Lymphocytes/immunology , Crohn Disease/immunology , Ileitis/immunology , Intestinal Mucosa/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Acridines/pharmacology , Adult , Animals , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Biological Transport , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeostasis/immunology , Humans , Ileitis/genetics , Ileitis/pathology , Ileum/immunology , Ileum/pathology , Immunity, Mucosal , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Oxidative Stress , Signal Transduction , Tetrahydroisoquinolines/pharmacology
15.
Mol Cell ; 65(6): 1122-1135.e5, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28306507

ABSTRACT

Human breast cancers that exhibit high proportions of immune cells and elevated levels of pro-inflammatory cytokines predict poor prognosis. Here, we demonstrate that treatment of human MCF-7 breast cancer cells with pro-inflammatory cytokines results in ERα-dependent activation of gene expression and proliferation, in the absence of ligand or presence of 4OH-tamoxifen (TOT). Cytokine activation of ERα and endocrine resistance is dependent on phosphorylation of ERα at S305 in the hinge domain. Phosphorylation of S305 by IKKß establishes an ERα cistrome that substantially overlaps with the estradiol (E2)-dependent ERα cistrome. Structural analyses suggest that S305-P forms a charge-linked bridge with the C-terminal F domain of ERα that enables inter-domain communication and constitutive activity from the N-terminal coactivator-binding site, revealing the structural basis of endocrine resistance. ERα therefore functions as a transcriptional effector of cytokine-induced IKKß signaling, suggesting a mechanism through which the tumor microenvironment controls tumor progression and endocrine resistance.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Cytokines/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor alpha/drug effects , Inflammation Mediators/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/analogs & derivatives , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Hep G2 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Interleukin-1beta/metabolism , MCF-7 Cells , Molecular Dynamics Simulation , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Phosphorylation , Protein Conformation , RNA Interference , Signal Transduction/drug effects , Structure-Activity Relationship , Tamoxifen/pharmacology , Transcription, Genetic , Transfection , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism
16.
Cell Chem Biol ; 24(1): 35-45, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28042045

ABSTRACT

Environmental estrogens and anti-hormone therapies for breast cancer have diverse tissue- and signaling-pathway-selective outcomes, but how estrogen receptor alpha (ERα) mediates this phenotypic diversity is poorly understood. We implemented a statistical approach to allow unbiased, parallel analyses of multiple crystal structures, and identified subtle perturbations of ERα structure by different synthetic and environmental estrogens. Many of these perturbations were in the sub-Å range, within the noise of the individual structures, but contributed significantly to the activities of synthetic and environmental estrogens. Combining structural perturbation data from many structures with quantitative cellular activity profiles of the ligands enabled identification of structural rules for ligand-specific allosteric signaling-predicting activity from structure. This approach provides a framework for understanding the diverse effects of environmental estrogens and for guiding iterative medicinal chemistry efforts to generate improved breast cancer therapies, an approach that can be applied to understanding other ligand-regulated allosteric signaling pathways.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogens/metabolism , Antineoplastic Agents, Hormonal/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Crystallography, X-Ray , Dimerization , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Estrogen Antagonists/chemistry , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Female , Humans , Ligands , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Tumor Cells, Cultured
17.
Proc Natl Acad Sci U S A ; 113(34): 9539-44, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27503891

ABSTRACT

The main cause of death globally remains debilitating heart conditions, such as dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM), which are often due to mutations of specific components of adhesion complexes. Vinculin regulates these complexes and plays essential roles in intercalated discs that are necessary for muscle cell function and coordinated movement and in the development and function of the heart. Humans bearing familial or sporadic mutations in vinculin suffer from chronic, progressively debilitating DCM that ultimately leads to cardiac failure and death, whereas autosomal dominant mutations in vinculin can also provoke HCM, causing acute cardiac failure. The DCM/HCM-associated mutants of vinculin occur in the 68-residue insert unique to the muscle-specific, alternatively spliced isoform of vinculin, termed metavinculin (MV). Contrary to studies that suggested that phosphoinositol-4,5-bisphosphate (PIP2) only induces vinculin homodimers, which are asymmetric, we show that phospholipid binding results in a domain-swapped symmetric MV dimer via a quasi-equivalent interface compared with vinculin involving R975. Although one of the two PIP2 binding sites is preserved, the symmetric MV dimer that bridges two PIP2 molecules differs from the asymmetric vinculin dimer that bridges only one PIP2 Unlike vinculin, wild-type MV and the DCM/HCM-associated R975W mutant bind PIP2 in their inactive conformations, and R975W MV fails to dimerize. Mutating selective vinculin residues to their corresponding MV residues, or vice versa, switches the isoform's dimeric constellation and lipid binding site. Collectively, our data suggest that MV homodimerization modulates microfilament attachment at muscular adhesion sites and furthers our understanding of MV-mediated cardiac remodeling.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate/chemistry , Vinculin/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Models, Molecular , Mutation , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thermodynamics , Vinculin/genetics , Vinculin/metabolism
18.
Biochemistry ; 54(17): 2758-68, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25880222

ABSTRACT

Vinculin localizes to cellular adhesions where it regulates motility, migration, development, wound healing, and response to force. Importantly, vinculin loss results in cancer phenotypes, cardiovascular disease, and embryonic lethality. At the plasma cell membrane, the most abundant phosphoinositide, phosphatidylinositol 4,5-bisphosphate (PIP2), binds the vinculin tail domain, Vt, and triggers homotypic and heterotypic interactions that amplify binding of vinculin to the actin network. Binding of PIP2 to Vt is necessary for maintaining optimal focal adhesions, for organizing stress fibers, for cell migration and spreading, and for the control of vinculin dynamics and turnover of focal adhesions. While the recently determined Vt/PIP2 crystal structure revealed the conformational changes occurring upon lipid binding and oligomerization, characterization of PIP2-induced vinculin oligomerization has been challenging in the adhesion biology field. Here, via a series of novel biochemical assays not performed in previous studies that relied on chemical cross-linking, we characterize the PIP2-induced vinculin oligomerization. Our results show that Vt/PIP2 forms a tight dimer with Vt or with the muscle-specific vinculin isoform, metavinculin, at sites of adhesion at the cell membrane. Insight into how PIP2 regulates clustering and into mechanisms that regulate cell adhesion allows the development for a more definite sensor for PIP2, and our developed techniques can be applied generally and thus open the door for the characterization of many other protein/PIP2 complexes under physiological conditions.


Subject(s)
Lipids/chemistry , Vinculin/chemistry , Cell Adhesion , Cell Membrane/chemistry , Chromatography, Gel , Dimerization , Surface Plasmon Resonance , Vinculin/metabolism
19.
J Cell Biol ; 207(5): 643-56, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25488920

ABSTRACT

Adherens junctions (AJs) and focal adhesion (FA) complexes are necessary for cell migration and morphogenesis, and for the development, growth, and survival of all metazoans. Vinculin is an essential regulator of both AJs and FAs, where it provides links to the actin cytoskeleton. Phosphatidylinositol 4,5-bisphosphate (PIP2) affects the functions of many targets, including vinculin. Here we report the crystal structure of vinculin in complex with PIP2, which revealed that PIP2 binding alters vinculin structure to direct higher-order oligomerization and suggests that PIP2 and F-actin binding to vinculin are mutually permissive. Forced expression of PIP2-binding-deficient mutants of vinculin in vinculin-null mouse embryonic fibroblasts revealed that PIP2 binding is necessary for maintaining optimal FAs, for organization of actin stress fibers, and for cell migration and spreading. Finally, photobleaching experiments indicated that PIP2 binding is required for the control of vinculin dynamics and turnover in FAs. Thus, through oligomerization, PIP2 directs a transient vinculin sequestration at FAs that is necessary for proper FA function.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate/chemistry , Vinculin/chemistry , Amino Acid Substitution , Animals , Binding Sites , Cells, Cultured , Crystallography, X-Ray , Focal Adhesions/physiology , Humans , Hydrogen Bonding , Mice , Models, Molecular , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Vinculin/physiology
20.
Nat Struct Mol Biol ; 20(2): 188-93, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23292143

ABSTRACT

The F-actin-binding cytoskeletal protein α-catenin interacts with ß-catenin-cadherin complexes and stabilizes cell-cell junctions. The ß-catenin-α-catenin complex cannot bind F-actin, whereas interactions of α-catenin with the cytoskeletal protein vinculin appear to be necessary to stabilize adherens junctions. Here we report the crystal structure of nearly full-length human α-catenin at 3.7-Å resolution. α-catenin forms an asymmetric dimer where the four-helix bundle domains of each subunit engage in distinct intermolecular interactions. This results in a left handshake-like dimer, wherein the two subunits have remarkably different conformations. The crystal structure explains why dimeric α-catenin has a higher affinity for F-actin than does monomeric α-catenin, why the ß-catenin-α-catenin complex does not bind F-actin, how activated vinculin links the cadherin-catenin complex to the cytoskeleton and why α-catenin but not inactive vinculin can bind F-actin.


Subject(s)
Adherens Junctions/metabolism , Models, Molecular , Protein Conformation , Vinculin/metabolism , alpha Catenin/chemistry , alpha Catenin/metabolism , Actins/metabolism , Adherens Junctions/genetics , Chromatography, Gel , Crystallography, X-Ray , Dimerization , Electrophoretic Mobility Shift Assay , Humans , Immunoblotting , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...