Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 6(57)2021 03 26.
Article in English | MEDLINE | ID: mdl-33771888

ABSTRACT

Regulatory T cells (Tregs) that promote tumor immune evasion are enriched in certain tumors and correlate with poor prognosis. However, mechanisms for Treg enrichment remain incompletely understood. We described a mechanism for Treg enrichment in mouse and human tumors mediated by the αvß8 integrin. Tumor cell αvß8 bound to latent transforming growth factor-ß (L-TGF-ß) presented on the surface of T cells, resulting in TGF-ß activation and immunosuppressive Treg differentiation in vitro. In vivo, tumor cell αvß8 expression correlated with Treg enrichment, immunosuppressive Treg gene expression, and increased tumor growth, which was reduced in mice by αvß8 inhibition or Treg depletion. Structural modeling and cell-based studies suggested a highly geometrically constrained complex forming between αvß8-expressing tumor cells and L-TGF-ß-expressing T cells, facilitating TGF-ß activation, independent of release and diffusion, and providing limited access to TGF-ß inhibitors. These findings suggest a highly localized tumor-specific mechanism for Treg enrichment.


Subject(s)
Integrins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Escape , Animals , Biomarkers , Cell Line, Tumor , Computational Biology/methods , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Humans , Mice , Models, Biological , Neoplasms/genetics , Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome
2.
Cell ; 180(3): 490-501.e16, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31955848

ABSTRACT

Integrin αvß8 binds with exquisite specificity to latent transforming growth factor-ß (L-TGF-ß). This binding is essential for activating L-TGF-ß presented by a variety of cell types. Inhibiting αvß8-mediated TGF-ß activation blocks immunosuppressive regulatory T cell differentiation, which is a potential therapeutic strategy in cancer. Using cryo-electron microscopy, structure-guided mutagenesis, and cell-based assays, we reveal the binding interactions between the entire αvß8 ectodomain and its intact natural ligand, L-TGF-ß, as well as two different inhibitory antibody fragments to understand the structural underpinnings of αvß8 binding specificity and TGF-ß activation. Our studies reveal a mechanism of TGF-ß activation where mature TGF-ß signals within the confines of L-TGF-ß and the release and diffusion of TGF-ß are not required. The structural details of this mechanism provide a rational basis for therapeutic strategies to inhibit αvß8-mediated L-TGF-ß activation.


Subject(s)
Cryoelectron Microscopy/methods , Integrins/chemistry , Integrins/metabolism , Latent TGF-beta Binding Proteins/chemistry , Latent TGF-beta Binding Proteins/metabolism , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/metabolism , Animals , Antibodies/immunology , Binding Sites , Bronchi/cytology , CHO Cells , Cricetulus , Female , Humans , Immunoglobulin Fab Fragments/immunology , Integrins/immunology , Lymphocyte Activation , Male , Mink , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , T-Lymphocytes, Regulatory/immunology
3.
JCI Insight ; 3(20)2018 10 18.
Article in English | MEDLINE | ID: mdl-30333313

ABSTRACT

TGF-ß is a promising immunotherapeutic target. It is expressed ubiquitously in a latent form that must be activated to function. Determination of where and how latent TGF-ß (L-TGF-ß) is activated in the tumor microenvironment could facilitate cell- and mechanism-specific approaches to immunotherapeutically target TGF-ß. Binding of L-TGF-ß to integrin αvß8 results in activation of TGF-ß. We engineered and used αvß8 antibodies optimized for blocking or detection, which - respectively - inhibit tumor growth in syngeneic tumor models or sensitively and specifically detect ß8 in human tumors. Inhibition of αvß8 potentiates cytotoxic T cell responses and recruitment of immune cells to tumor centers - effects that are independent of PD-1/PD-L1. ß8 is expressed on the cell surface at high levels by tumor cells, not immune cells, while the reverse is true of L-TGF-ß, suggesting that tumor cell αvß8 serves as a platform for activating cell-surface L-TGF-ß presented by immune cells. Transcriptome analysis of tumor-associated lymphoid cells reveals macrophages as a key cell type responsive to ß8 inhibition with major increases in chemokine and tumor-eliminating genes. High ß8 expression in tumor cells is seen in 20%-80% of various cancers, which rarely coincides with high PD-L1 expression. These data suggest tumor cell αvß8 is a PD-1/PD-L1-independent immunotherapeutic target.


Subject(s)
Integrins/metabolism , Macrophages/immunology , Neoplasms/immunology , Transforming Growth Factor beta/metabolism , Tumor Escape/immunology , Animals , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Cell Line, Tumor , Computer Simulation , Disease Models, Animal , Female , Humans , Integrins/antagonists & inhibitors , Kaplan-Meier Estimate , Macrophages/metabolism , Male , Mice , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/mortality , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Escape/drug effects , Tumor Microenvironment/immunology
4.
Nat Struct Mol Biol ; 25(8): 698-704, 2018 08.
Article in English | MEDLINE | ID: mdl-30061598

ABSTRACT

Integrins are conformationally flexible cell surface receptors that survey the extracellular environment for their cognate ligands. Interactions with ligands are thought to be linked to global structural rearrangements involving transitions between bent, extended-closed and extended-open forms. Thus far, structural details are lacking for integrins in the extended conformations due to extensive flexibility between the headpiece and legs in this conformation. Here we present single-particle electron cryomicroscopy structures of human αvß8 integrin in the extended-closed conformation, which has been considered to be a low-affinity intermediate. Our structures show the headpiece rotating about a flexible αv knee, suggesting a ligand surveillance mechanism for integrins in their extended-closed form. Our model predicts that the extended conformation is mainly stabilized by an interface formed between flexible loops in the upper and lower domains of the αv leg. Confirming these findings with the αvß3 integrin suggests that our model of stabilizing the extended-closed conformation is generalizable to other integrins.


Subject(s)
Cryoelectron Microscopy/methods , Integrins/metabolism , Amino Acid Sequence , Humans , Integrins/chemistry , Protein Conformation , Sequence Homology, Amino Acid
5.
Toxins (Basel) ; 10(2)2018 02 15.
Article in English | MEDLINE | ID: mdl-29462889

ABSTRACT

The standard of treatment for botulism, equine antitoxin, is a foreign protein with associated safety issues and a short serum half-life which excludes its use as a prophylactic antitoxin and makes it a less-than-optimal therapeutic. Due to these limitations, a recombinant monoclonal antibody (mAb) product is preferable. It has been shown that combining three mAbs that bind non-overlapping epitopes leads to highly potent botulinum neurotoxin (BoNT) neutralization. Recently, a triple human antibody combination for BoNT/A has demonstrated potent toxin neutralization in mouse models with no serious adverse events when tested in a Phase I clinical trial. However, a triple antibody therapeutic poses unique development and manufacturing challenges. Thus, potentially to streamline development of BoNT antitoxins, we sought to achieve the potency of multiple mAb combinations in a single IgG-based molecule that has a long serum half-life. The design, production, and testing of a single tri-epitopic IgG1-based mAb (TeAb) containing the binding sites of each of the three parental BoNT/A mAbs yielded an antibody of nearly equal potency to the combination. The approach taken here could be applied to the design and creation of other multivalent antibodies that could be used for a variety of applications, including toxin elimination.


Subject(s)
Antibodies, Monoclonal/immunology , Botulinum Toxins, Type A/immunology , Epitopes/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/pharmacology , CHO Cells , Cricetulus , Female , Mice , Neurons/metabolism , Neutralization Tests , Rats
6.
Sci Transl Med ; 6(241): 241ra79, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24944194

ABSTRACT

Airway remodeling, caused by inflammation and fibrosis, is a major component of chronic obstructive pulmonary disease (COPD) and currently has no effective treatment. Transforming growth factor-ß (TGF-ß) has been widely implicated in the pathogenesis of airway remodeling in COPD. TGF-ß is expressed in a latent form that requires activation. The integrin αvß8 (encoded by the itgb8 gene) is a receptor for latent TGF-ß and is essential for its activation. Expression of integrin αvß8 is increased in airway fibroblasts in COPD and thus is an attractive therapeutic target for the treatment of airway remodeling in COPD. We demonstrate that an engineered optimized antibody to human αvß8 (B5) inhibited TGF-ß activation in transgenic mice expressing only human and not mouse ITGB8. The B5 engineered antibody blocked fibroinflammatory responses induced by tobacco smoke, cytokines, and allergens by inhibiting TGF-ß activation. To clarify the mechanism of action of B5, we used hydrodynamic, mutational, and electron microscopic methods to demonstrate that αvß8 predominantly adopts a constitutively active, extended-closed headpiece conformation. Epitope mapping and functional characterization of B5 revealed an allosteric mechanism of action due to locking-in of a low-affinity αvß8 conformation. Collectively, these data demonstrate a new model for integrin function and present a strategy to selectively target the TGF-ß pathway to treat fibroinflammatory airway diseases.


Subject(s)
Tracheitis/therapy , Transforming Growth Factor beta/metabolism , Animals , Humans , Mice , Mice, Transgenic
7.
J Biol Chem ; 288(32): 23203-11, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23814063

ABSTRACT

The chromosomal passenger complex (CPC) is a key regulator of mitosis in eukaryotes. It comprises four essential and conserved proteins known in mammals/yeasts as Aurora B/Ipl1, INCENP/Sli15, Survivin/Bir1, and Borealin/Nbl1. These subunits act together in a highly controlled fashion. Regulation of Aurora B/Ipl1 kinase activity and localization is critical for CPC function. Although regulation of CPC localization and kinase activity in vivo has been investigated elsewhere, studies on the complete, four-subunit CPC and its basic biochemical properties are only beginning. Here we describe the biochemical characterization of purified and complete Saccharomyces cerevisiae four-subunit CPC. We determined the affinity of the CPC for microtubules and demonstrated that the binding of CPC to microtubules is primarily electrostatic in nature and depends on the acidic C-terminal tail (E-hook) of tubulin. Moreover, phosphorylation of INCENP/Sli15 on its microtubule binding region also negatively regulates CPC affinity for microtubules. Furthermore, we show that phosphorylation of INCENP/Sli15 is required for activation of the kinase Aurora B/Ipl1 and can occur in trans. Although phosphorylation of INCENP/Sli15 is essential for activation, we determined that a version of the CPC lacking the INCENP/Sli15 microtubule binding region (residues Glu-91 to Ile-631) is able to form an intact complex that retains microtubule binding activity. Thus, we conclude that this INCENP/Sli15 linker domain plays a largely regulatory function and is not essential for complex formation or microtubule binding.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Multiprotein Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Aurora Kinases , Intracellular Signaling Peptides and Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Multiprotein Complexes/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
8.
J Cell Biol ; 194(1): 137-53, 2011 Jul 11.
Article in English | MEDLINE | ID: mdl-21727193

ABSTRACT

Dynamic microtubules facilitate chromosome arrangement before anaphase, whereas during anaphase microtubule stability assists chromosome separation. Changes in microtubule dynamics at the metaphase-anaphase transition are regulated by Cdk1. Cdk1-mediated phosphorylation of Sli15/INCENP promotes preanaphase microtubule dynamics by preventing chromosomal passenger complex (CPC; Sli15/INCENP, Bir1/Survivin, Nbl1/Borealin, Ipl1/Aurora) association with spindles. However, whether Cdk1 has sole control over microtubule dynamics, and how CPC-microtubule association influences microtubule behavior, are unclear. Here, we show that Ipl1/Aurora-dependent phosphorylation of Sli15/INCENP modulates microtubule dynamics by preventing CPC binding to the preanaphase spindle and to the central spindle until late anaphase, facilitating spatiotemporal control of microtubule dynamics required for proper metaphase centromere positioning and anaphase spindle elongation. Decreased Ipl1-dependent Sli15 phosphorylation drives direct CPC binding to microtubules, revealing how the CPC influences microtubule dynamics. We propose that Cdk1 and Ipl1/Aurora cooperatively modulate microtubule dynamics and that Ipl1/Aurora-dependent phosphorylation of Sli15 controls spindle function by excluding the CPC from spindle regions engaged in microtubule polymerization.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Spindle Apparatus/metabolism , Aurora Kinases , Phosphorylation
9.
Methods Cell Biol ; 95: 373-90, 2010.
Article in English | MEDLINE | ID: mdl-20466145

ABSTRACT

Vinca domain ligands are small molecules that interfere with the binding of vinblastine to tubulin and inhibit microtubule assembly. Many such compounds cause isodesmic association which results in difficulties in biochemical or structural studies of their interaction with tubulin. The complex of two tubulins with the stathmin-like domain of the RB3 protein (T(2)R) is a protofilament-like short assembly that does not assemble further. This has allowed structural studies of the binding of several vinca domain ligands by X-ray crystallography as crystals of the corresponding complexes diffract to near atomic resolution. This proved that their sites are located at the interface of two tubulin molecules arranged as in a curved protofilament. These sites overlap with that of vinblastine. Structural data are generally consistent with the results of available structure-function studies, though subtle differences exist. Binding in solution to the vinca domain displayed in T(2)R is conveniently studied by fluorescence spectroscopy or by monitoring inhibition of the T(2)R GTPase activity. In addition, inhibition of nucleotide exchange allows characterization of the binding to the vinca domain moiety displayed by the beta-subunit of an isolated tubulin molecule. T(2)R is therefore a useful tool to characterize and dissect the binding of vinca domain ligands to tubulin. In addition, these studies have provided new information on the interaction of tubulin with guanine nucleotides, namely on the mechanisms of nucleotide exchange and hydrolysis.


Subject(s)
Tubulin/chemistry , Tubulin/metabolism , Vinca Alkaloids/metabolism , Animals , Biochemistry/methods , Fluorescent Dyes/pharmacology , Humans , Models, Biological , Models, Molecular , Protein Binding/physiology , Protein Conformation/drug effects , Structure-Activity Relationship , Vinca Alkaloids/pharmacokinetics
10.
J Biol Chem ; 284(11): 6909-17, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19131341

ABSTRACT

Microtubules are cytoskeletal components involved in multiple cell functions such as mitosis, motility, or intracellular traffic. In vivo, these polymers made of alphabeta-tubulin nucleate mostly from the centrosome to establish the interphasic microtubule network or, during mitosis, the mitotic spindle. Centrosomal P4.1-associated protein (CPAP; also named CENPJ) is a centrosomal protein involved in the assembly of centrioles and important for the centrosome function. This protein contains a microtubule-destabilizing region referred to as PN2-3. Here we decrypt the microtubule destabilization activity of PN2-3 at the molecular level and show that it results from the sequestration of tubulin by PN2-3 in a non-polymerizable 1:1 complex. We also map the tubulin/PN2-3 interaction both on the PN2-3 sequence and on the tubulin surface. NMR and CD data on free PN2-3 in solution show that this is an intrinsically unstructured protein that comprises a 23-amino acid residue alpha-helix. This helix is embedded in a 76-residue region that interacts strongly with tubulin. The interference of PN2-3 with well characterized tubulin properties, namely GTPase activity, nucleotide exchange, vinblastine-induced self-assembly, and stathmin family protein binding, highlights the beta subunit surface located at the intermolecular longitudinal interface when tubulin is embedded in a microtubule as a tubulin/PN2-3 interaction area. These findings characterize the PN2-3 fragment of CPAP as a protein with an unprecedented tubulin sequestering mechanism distinct from that of stathmin family proteins.


Subject(s)
Centrioles/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism , Animals , Circular Dichroism , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary/physiology , Sheep
11.
J Med Chem ; 52(1): 134-42, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-19072542

ABSTRACT

Ten hybrids of vinca alkaloids and phomopsin A have been synthesized by linking the octahydrophomopsin lateral chain to the tertiary amine of the cleavamine moiety of anhydrovinblastine (AVLB) and vinorelbine. These compounds have been elaborated in order to obtain original products that may interfere with both binding sites of vinblastine (VLB) and phomopsin in tubulin. Although NMR and molecular modeling studies have shown that the orientation of the added peptide chains of these hybrids is not the same as those of phomopsin A, most of them are very potent inhibitors of microtubules assembly and they present good cytotoxicity against KB cell line. These interesting biological activities may eventually be explained by the fact that their lateral chain resides in a pocket distinct from that of the phomopsin A peptide, at the interface of tubulins beta and alpha.


Subject(s)
Antimitotic Agents/chemical synthesis , Mycotoxins/chemical synthesis , Mycotoxins/pharmacology , Tubulin Modulators/chemical synthesis , Vinca Alkaloids/chemical synthesis , Vinca Alkaloids/pharmacology , Antimitotic Agents/chemistry , Antimitotic Agents/pharmacology , Cell Death/drug effects , Cell Line , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Microtubules/drug effects , Microtubules/metabolism , Molecular Structure , Mycotoxins/chemistry , Mycotoxins/toxicity , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Tubulin Modulators/toxicity , Vinblastine/analogs & derivatives , Vinblastine/chemistry , Vinca Alkaloids/chemistry , Vinca Alkaloids/toxicity , Vinorelbine
12.
EMBO Rep ; 9(11): 1101-6, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18787557

ABSTRACT

The tubulin vinca domain is the target of widely different microtubule inhibitors that interfere with the binding of vinblastine. Although all these ligands inhibit the hydrolysis of GTP, they affect nucleotide exchange to variable extents. The structures of two vinca domain antimitotic peptides--phomopsin A and soblidotin (a dolastatin 10 analogue)--bound to tubulin in a complex with a stathmin-like domain show that their sites partly overlap with that of vinblastine and extend the definition of the vinca domain. The structural data, together with the biochemical results from the ligands we studied, highlight two main contributors in nucleotide exchange: the flexibility of the tubulin subunits' arrangement at their interfaces and the residues in the carboxy-terminal part of the beta-tubulin H6-H7 loop. The structures also highlight common features of the mechanisms by which vinca domain ligands favour curved tubulin assemblies and destabilize microtubules.


Subject(s)
Tubulin/metabolism , Ligands , Microtubules/metabolism , Models, Molecular , Mycotoxins/metabolism , Oligopeptides/metabolism , Protein Structure, Tertiary , Tubulin/chemistry , Tubulin/pharmacology , Vinblastine/metabolism
13.
Biochemistry ; 46(37): 10595-602, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17711308

ABSTRACT

Microtubules are dynamically unstable tubulin polymers that interconvert stochastically between growing and shrinking states, a property central to their cellular functions. Following its incorporation in microtubules, tubulin hydrolyzes one GTP molecule. Microtubule dynamic instability depends on GTP hydrolysis so that this activity is crucial to the regulation of microtubule assembly. Tubulin also has a much lower GTPase activity in solution. We have used ternary complexes made of two tubulin molecules and one stathmin-like domain to investigate the mechanism of the tubulin GTPase activity in solution. We show that whereas stathmin-like domains and colchicine enhance this activity, it is inhibited by vinblastine and by the N-terminal part of stathmin-like domains. Taken together with the structures of the tubulin-colchicine-stathmin-like domain-vinblastine complex and of microtubules, our results lead to the conclusions that the tubulin-colchicine GTPase activity in solution is caused by tubulin-tubulin associations and that the residues involved in catalysis comprise the beta tubulin GTP binding site and alpha tubulin residues that participate in intermolecular interactions in protofilaments. This site resembles the one that has been proposed to give rise to GTP hydrolysis in microtubules. The widely different hydrolysis rates in these two sites result at least in part from the curved and straight tubulin assemblies in solution and in microtubules, respectively.


Subject(s)
GTP Phosphohydrolases/metabolism , Stathmin/chemistry , Tubulin/metabolism , Animals , Cattle , Colchicine/chemistry , Colchicine/metabolism , Ligands , Podophyllotoxin/chemistry , Podophyllotoxin/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Vinblastine/chemistry , Vinblastine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...