Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 16(755): eadg7123, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38985855

ABSTRACT

Two types of engineered T cells have been successfully used to treat patients with cancer, one with an antigen recognition domain derived from antibodies [chimeric antigen receptors (CARs)] and the other derived from T cell receptors (TCRs). CARs use high-affinity antigen-binding domains and costimulatory domains to induce T cell activation but can only react against target cells with relatively high amounts of antigen. TCRs have a much lower affinity for their antigens but can react against target cells displaying only a few antigen molecules. Here, we describe a new type of receptor, called a Co-STAR (for costimulatory synthetic TCR and antigen receptor), that combines aspects of both CARs and TCRs. In Co-STARs, the antigen-recognizing components of TCRs are replaced by high-affinity antibody fragments, and costimulation is provided by two modules that drive NF-κB signaling (MyD88 and CD40). Using a TCR-mimic antibody fragment that targets a recurrent p53 neoantigen presented in a common human leukocyte antigen (HLA) allele, we demonstrate that T cells equipped with Co-STARs can kill cancer cells bearing low densities of antigen better than T cells engineered with conventional CARs and patient-derived TCRs in vitro. In mouse models, we show that Co-STARs mediate more robust T cell expansion and more durable tumor regressions than TCRs similarly modified with MyD88 and CD40 costimulation. Our data suggest that Co-STARs may have utility for other peptide-HLA antigens in cancer and other targets where antigen density may limit the efficacy of engineered T cells.


Subject(s)
Neoplasms , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Humans , Animals , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Neoplasms/immunology , Neoplasms/therapy , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Signal Transduction
2.
Nat Rev Cancer ; 24(6): 399-426, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38740967

ABSTRACT

The greatest challenge in cancer therapy is to eradicate cancer cells with minimal damage to normal cells. Targeted therapy has been developed to meet that challenge, showing a substantially increased therapeutic index compared with conventional cancer therapies. Antibodies are important members of the family of targeted therapeutic agents because of their extraordinarily high specificity to the target antigens. Therapeutic antibodies use a range of mechanisms that directly or indirectly kill the cancer cells. Early antibodies were developed to directly antagonize targets on cancer cells. This was followed by advancements in linker technologies that allowed the production of antibody-drug conjugates (ADCs) that guide cytotoxic payloads to the cancer cells. Improvement in our understanding of the biology of T cells led to the production of immune checkpoint-inhibiting antibodies that indirectly kill the cancer cells through activation of the T cells. Even more recently, bispecific antibodies were synthetically designed to redirect the T cells of a patient to kill the cancer cells. In this Review, we summarize the different approaches used by therapeutic antibodies to target cancer cells. We discuss their mechanisms of action, the structural basis for target specificity, clinical applications and the ongoing research to improve efficacy and reduce toxicity.


Subject(s)
Immunoconjugates , Neoplasms , Humans , Neoplasms/immunology , Neoplasms/drug therapy , Immunoconjugates/therapeutic use , Immunoconjugates/pharmacology , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Animals , T-Lymphocytes/immunology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology
3.
J Biol Chem ; 300(3): 105715, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38309503

ABSTRACT

NEDD4L is a HECT-type E3 ligase that catalyzes the addition of ubiquitin to intracellular substrates such as the cardiac voltage-gated sodium channel, NaV1.5. The intramolecular interactions of NEDD4L regulate its enzymatic activity which is essential for proteostasis. For NaV1.5, this process is critical as alterations in Na+ current is involved in cardiac diseases including arrhythmias and heart failure. In this study, we perform extensive biochemical and functional analyses that implicate the C2 domain and the first WW-linker (1,2-linker) in the autoregulatory mechanism of NEDD4L. Through in vitro and electrophysiological experiments, the NEDD4L 1,2-linker was determined to be important in substrate ubiquitination of NaV1.5. We establish the preferred sites of ubiquitination of NEDD4L to be in the second WW-linker (2,3-linker). Interestingly, NEDD4L ubiquitinates the cytoplasmic linker between the first and second transmembrane domains of the channel (DI-DII) of NaV1.5. Moreover, we design a genetically encoded modulator of Nav1.5 that achieves Na+ current reduction using the NEDD4L HECT domain as cargo of a NaV1.5-binding nanobody. These investigations elucidate the mechanisms regulating the NEDD4 family and furnish a new molecular framework for understanding NaV1.5 ubiquitination.


Subject(s)
Endosomal Sorting Complexes Required for Transport , NAV1.5 Voltage-Gated Sodium Channel , Nedd4 Ubiquitin Protein Ligases , Ubiquitination , Endosomal Sorting Complexes Required for Transport/metabolism , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Ubiquitin/metabolism , Humans , NAV1.5 Voltage-Gated Sodium Channel/metabolism , HEK293 Cells
4.
ACS Bio Med Chem Au ; 3(6): 516-527, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38144259

ABSTRACT

NaV1.7, the neuronal voltage-gated sodium channel isoform, plays an important role in the human body's ability to feel pain. Mutations within NaV1.7 have been linked to pain-related syndromes, such as insensitivity to pain. To date, the regulation and internalization mechanisms of the NaV1.7 channel are not well known at a biochemical level. In this study, we perform biochemical and biophysical analyses that establish that the HECT-type E3 ligase, NEDD4L, ubiquitinates the cytoplasmic C-terminal (CT) region of NaV1.7. Through in vitro ubiquitination and mass spectrometry experiments, we identify, for the first time, the lysine residues of NaV1.7 within the CT region that get ubiquitinated. Furthermore, binding studies with an NEDD4L E3 ligase modulator (ubiquitin variant) highlight the dynamic partnership between NEDD4L and NaV1.7. These investigations provide a framework for understanding how NEDD4L-dependent regulation of the channel can influence the NaV1.7 function.

5.
Nat Commun ; 14(1): 5063, 2023 08 21.
Article in English | MEDLINE | ID: mdl-37604828

ABSTRACT

Specificity remains a major challenge to current therapeutic strategies for cancer. Mutation associated neoantigens (MANAs) are products of genetic alterations, making them highly specific therapeutic targets. MANAs are HLA-presented (pHLA) peptides derived from intracellular mutant proteins that are otherwise inaccessible to antibody-based therapeutics. Here, we describe the cryo-EM structure of an antibody-MANA pHLA complex. Specifically, we determine a TCR mimic (TCRm) antibody bound to its MANA target, the KRASG12V peptide presented by HLA-A*03:01. Hydrophobic residues appear to account for the specificity of the mutant G12V residue. We also determine the structure of the wild-type G12 peptide bound to HLA-A*03:01, using X-ray crystallography. Based on these structures, we perform screens to validate the key residues required for peptide specificity. These experiments led us to a model for discrimination between the mutant and the wild-type peptides presented on HLA-A*03:01 based exclusively on hydrophobic interactions.


Subject(s)
Antibodies , Proto-Oncogene Proteins p21(ras) , Proto-Oncogene Proteins p21(ras)/genetics , Recognition, Psychology , Hydrophobic and Hydrophilic Interactions , HLA-A Antigens/genetics
6.
Biochemistry ; 62(5): 1012-1025, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36820504

ABSTRACT

The MID1 TRIM protein is important for ventral midline development in vertebrates, and mutations of its B-box1 domain result in several birth defects. The B-box1 domain of the human MID1 protein binds two zinc atoms and adopt a similar ßßα-RING structure. This domain is required for the efficient ubiquitination of protein phosphatase 2A, alpha4, and fused kinase. Considering the structural similarity, the MID1 B-box1 domain exhibits mono-autoubiquitination activity, in contrast to poly-autoubiquitination observed for RING E3 ligases. To understand its mechanism of action, the interaction of the B-box1 domain with Ube2D1 (UbcH5a, E2), a preferred E2 ligase, is investigated. Using isothermal titration calorimetry, the MID1 RING and B-box1 domains were observed to have similar binding affinities with the Ube2D1 protein. However, NMR 15N-1H Heteronuclear Single Quantum Coherence titration, 15N relaxation data, and High Ambiguity Driven protein-protein DOCKing (HADDOCK) calculations show the B-box1 domain binding on a surface distinct from where RING domains bind. The novel binding interaction shows the B-box1 domain partially overlapping the noncovalent Ube2D1 and a ubiquitin binding site that is necessary for poly-autoubiquitination activity. The B-box1 domain also displaces the ubiquitin from the Ube2D1 protein. These studies reveal a novel binding interaction between the zinc-binding ßßα-fold B-box1 domain and the Ube2D enzyme family and that this difference in binding, compared to RING E3 ligases, provides a rationale for its auto-monoubiquitination E3 ligase activity.


Subject(s)
Microtubule Proteins , Transcription Factors , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases , Humans , Amino Acid Sequence , Microtubule Proteins/chemistry , Models, Molecular , Protein Structure, Tertiary , Transcription Factors/chemistry , Ubiquitin/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Protein Ligases/chemistry , Ubiquitination , Zinc/chemistry
7.
Nat Commun ; 14(1): 17, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36596784

ABSTRACT

The therapeutic applications of antibodies are manifold and the emergence of SARS-CoV-2 provides a cogent example of the value of rapidly identifying biologically active antibodies. We describe an approach called SLISY (Sequencing-Linked ImmunoSorbent assaY) that in a single experiment can assess the binding specificity of millions of clones, be applied to any screen that links DNA sequence to a potential binding moiety, and requires only a single round of biopanning. We demonstrate this approach using an scFv library applied to cellular and protein targets to identify specific or broadly reacting antibodies. For a cellular target, we use paired HLA knockout cell lines to identify a panel of antibodies specific to HLA-A3. For a protein target, SLISY identifies 1279 clones that bound to the Receptor Binding Domain of the SARS-CoV-2 spike protein, with >40% of tested clones also neutralizing its interaction with ACE2 in in vitro assays. Using a multi-comparison SLISY against the Beta, Gamma, and Delta variants, we recovered clones that exhibited broad-spectrum neutralizing potential in vitro. By evaluating millions of scFvs simultaneously against multiple targets, SLISY allows the rapid identification of candidate scFvs with defined binding profiles facilitating the identification of antibodies with the desired biological activity.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing , Antibodies, Viral
8.
J Biol Chem ; 298(4): 101763, 2022 04.
Article in English | MEDLINE | ID: mdl-35202650

ABSTRACT

Voltage-gated sodium channels, NaVs, are responsible for the rapid rise of action potentials in excitable tissues. NaV channel mutations have been implicated in several human genetic diseases, such as hypokalemic periodic paralysis, myotonia, and long-QT and Brugada syndromes. Here, we generated high-affinity anti-NaV nanobodies (Nbs), Nb17 and Nb82, that recognize the NaV1.4 (skeletal muscle) and NaV1.5 (cardiac muscle) channel isoforms. These Nbs were raised in llama (Lama glama) and selected from a phage display library for high affinity to the C-terminal (CT) region of NaV1.4. The Nbs were expressed in Escherichia coli, purified, and biophysically characterized. Development of high-affinity Nbs specifically targeting a given human NaV isoform has been challenging because they usually show undesired crossreactivity for different NaV isoforms. Our results show, however, that Nb17 and Nb82 recognize the CTNaV1.4 or CTNaV1.5 over other CTNav isoforms. Kinetic experiments by biolayer interferometry determined that Nb17 and Nb82 bind to the CTNaV1.4 and CTNaV1.5 with high affinity (KD ∼ 40-60 nM). In addition, as proof of concept, we show that Nb82 could detect NaV1.4 and NaV1.5 channels in mammalian cells and tissues by Western blot. Furthermore, human embryonic kidney cells expressing holo NaV1.5 channels demonstrated a robust FRET-binding efficiency for Nb17 and Nb82. Our work lays the foundation for developing Nbs as anti-NaV reagents to capture NaVs from cell lysates and as molecular visualization agents for NaVs.


Subject(s)
Single-Domain Antibodies , Voltage-Gated Sodium Channels , Animals , Cells, Cultured , Escherichia coli/genetics , Humans , Long QT Syndrome/metabolism , Mammals/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single-Domain Antibodies/genetics , Single-Domain Antibodies/metabolism , Voltage-Gated Sodium Channels/genetics , Voltage-Gated Sodium Channels/metabolism
9.
ACS Chem Biol ; 17(1): 68-76, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34941261

ABSTRACT

Akt is a Ser/Thr protein kinase that regulates cell growth and metabolism and is considered a therapeutic target for cancer. Regulation of Akt by membrane recruitment and post-translational modifications (PTMs) has been extensively studied. The most well-established mechanism for cellular Akt activation involves phosphorylation on its activation loop on Thr308 by PDK1 and on its C-terminal tail on Ser473 by mTORC2. In addition, dual phosphorylation on Ser477 and Thr479 has been shown to activate Akt. Other C-terminal tail PTMs have been identified, but their functional impacts have not been well-characterized. Here, we investigate the regulatory effects of phosphorylation of Tyr474 and O-GlcNAcylation of Ser473 on Akt. We use expressed protein ligation as a tool to produce semisynthetic Akt proteins containing phosphoTyr474 and O-GlcNAcSer473 to dissect the enzymatic functions of these PTMs. We find that O-GlcNAcylation at Ser473 and phosphorylation at Tyr474 can also partially increase Akt's kinase activity toward both peptide and protein substrates. Additionally, we performed kinase assays employing human protein microarrays to investigate global substrate specificity of Akt, comparing phosphorylated versus O-GlcNAcylated Ser473 forms. We observed a high similarity in the protein substrates phosphorylated by phosphoSer473 Akt and O-GlcNAcSer473 Akt. Two Akt substrates identified using microarrays, PPM1H, a protein phosphatase, and NEDD4L, an E3 ubiquitin ligase, were validated in solution-phase assays and cell transfection experiments.


Subject(s)
Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Animals , HCT116 Cells , Humans , Insecta , Phosphorylation , Proto-Oncogene Proteins c-akt/chemical synthesis , Sf9 Cells
10.
Nat Commun ; 12(1): 5271, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489470

ABSTRACT

Chimeric antigen receptor (CAR) T cells have emerged as a promising class of therapeutic agents, generating remarkable responses in the clinic for a subset of human cancers. One major challenge precluding the wider implementation of CAR therapy is the paucity of tumor-specific antigens. Here, we describe the development of a CAR targeting the tumor-specific isocitrate dehydrogenase 2 (IDH2) with R140Q mutation presented on the cell surface in complex with a common human leukocyte antigen allele, HLA-B*07:02. Engineering of the hinge domain of the CAR, as well as crystal structure-guided optimization of the IDH2R140Q-HLA-B*07:02-targeting moiety, enhances the sensitivity and specificity of CARs to enable targeting of this HLA-restricted neoantigen. This approach thus holds promise for the development and optimization of immunotherapies specific to other cancer driver mutations that are difficult to target by conventional means.


Subject(s)
HLA-B7 Antigen/chemistry , Isocitrate Dehydrogenase/metabolism , Protein Engineering/methods , Receptors, Chimeric Antigen/chemistry , Animals , Antigens, Neoplasm/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Epitopes , HLA-B7 Antigen/metabolism , Humans , Immunoglobulin Fab Fragments/chemistry , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mutation , Peptide Library , Protein Conformation , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/physiology
11.
Sci Immunol ; 6(57)2021 03 01.
Article in English | MEDLINE | ID: mdl-33649101

ABSTRACT

Mutations in the RAS oncogenes occur in multiple cancers, and ways to target these mutations has been the subject of intense research for decades. Most of these efforts are focused on conventional small-molecule drugs rather than antibody-based therapies because the RAS proteins are intracellular. Peptides derived from recurrent RAS mutations, G12V and Q61H/L/R, are presented on cancer cells in the context of two common human leukocyte antigen (HLA) alleles, HLA-A3 and HLA-A1, respectively. Using phage display, we isolated single-chain variable fragments (scFvs) specific for each of these mutant peptide-HLA complexes. The scFvs did not recognize the peptides derived from the wild-type form of RAS proteins or other related peptides. We then sought to develop an immunotherapeutic agent that was capable of killing cells presenting very low levels of these RAS-derived peptide-HLA complexes. Among many variations of bispecific antibodies tested, one particular format, the single-chain diabody (scDb), exhibited superior reactivity to cells expressing low levels of neoantigens. We converted the scFvs to this scDb format and demonstrated that they were capable of inducing T cell activation and killing of target cancer cells expressing endogenous levels of the mutant RAS proteins and cognate HLA alleles. CRISPR-mediated alterations of the HLA and RAS genes provided strong genetic evidence for the specificity of the scDbs. Thus, this approach could be applied to other common oncogenic mutations that are difficult to target by conventional means, allowing for more specific anticancer therapeutics.


Subject(s)
Antibodies, Bispecific/pharmacology , Antigens, Neoplasm , Biomarkers, Tumor/antagonists & inhibitors , Mutant Proteins/antagonists & inhibitors , ras Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Bispecific/immunology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Cell Line , Cross Reactions , HLA Antigens/immunology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mutant Proteins/chemistry , Mutant Proteins/immunology , Mutation , Peptide Fragments , Protein Binding/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , ras Proteins/chemistry , ras Proteins/genetics , ras Proteins/immunology
12.
Science ; 371(6533)2021 03 05.
Article in English | MEDLINE | ID: mdl-33649166

ABSTRACT

TP53 (tumor protein p53) is the most commonly mutated cancer driver gene, but drugs that target mutant tumor suppressor genes, such as TP53, are not yet available. Here, we describe the identification of an antibody highly specific to the most common TP53 mutation (R175H, in which arginine at position 175 is replaced with histidine) in complex with a common human leukocyte antigen-A (HLA-A) allele on the cell surface. We describe the structural basis of this specificity and its conversion into an immunotherapeutic agent: a bispecific single-chain diabody. Despite the extremely low p53 peptide-HLA complex density on the cancer cell surface, the bispecific antibody effectively activated T cells to lyse cancer cells that presented the neoantigen in vitro and in mice. This approach could in theory be used to target cancers containing mutations that are difficult to target in conventional ways.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , HLA-A2 Antigen/immunology , Neoplasms/therapy , Tumor Suppressor Protein p53/immunology , Alleles , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/therapeutic use , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/therapeutic use , Arginine/genetics , COS Cells , Chlorocebus aethiops , Female , HEK293 Cells , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , Histidine/genetics , Humans , Immunization, Passive , Jurkat Cells , Lymphocyte Activation , Mice, Inbred NOD , Mutation , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
13.
Sci Transl Med ; 13(584)2021 03 10.
Article in English | MEDLINE | ID: mdl-33649188

ABSTRACT

Immunotherapies such as chimeric antigen receptor (CAR) T cells and bispecific antibodies redirect healthy T cells to kill cancer cells expressing the target antigen. The pan-B cell antigen-targeting immunotherapies have been remarkably successful in treating B cell malignancies. Such therapies also result in the near-complete loss of healthy B cells, but this depletion is well tolerated by patients. Although analogous targeting of pan-T cell markers could, in theory, help control T cell cancers, the concomitant healthy T cell depletion would result in severe and unacceptable immunosuppression. Thus, therapies directed against T cell cancers require more selective targeting. Here, we describe an approach to target T cell cancers through T cell receptor (TCR) antigens. Each T cell, normal or malignant, expresses a unique TCR ß chain generated from 1 of 30 TCR ß chain variable gene families (TRBV1 to TRBV30). We hypothesized that bispecific antibodies targeting a single TRBV family member expressed in malignant T cells could promote killing of these cancer cells, while preserving healthy T cells that express any of the other 29 possible TRBV family members. We addressed this hypothesis by demonstrating that bispecific antibodies targeting TRBV5-5 (α-V5) or TRBV12 (α-V12) specifically lyse relevant malignant T cell lines and patient-derived T cell leukemias in vitro. Treatment with these antibodies also resulted in major tumor regressions in mouse models of human T cell cancers. This approach provides an off-the-shelf, T cell cancer selective targeting approach that preserves enough healthy T cells to maintain cellular immunity.


Subject(s)
Antibodies, Bispecific , Lymphoproliferative Disorders/therapy , T-Lymphocytes/pathology , Humans , Receptors, Antigen, T-Cell, alpha-beta
14.
FEBS J ; 284(14): 2183-2193, 2017 07.
Article in English | MEDLINE | ID: mdl-28548391

ABSTRACT

Mutations of human MID1 are associated with X-linked Opitz G Syndrome (XLOS), which is characterized by midline birth defects. XLOS-observed mutations within the MID1 B-box1 domain are associated with cleft lip/palate, wide-spaced eyes and hyperspadias. Three of the four XLOS-observed mutations in the B-box1 domain results in unfolding but the structural and functional effects of the P151L mutation is not characterized. Here, we demonstrate that the P151L mutation does not disrupt the overall tertiary structure of the B-box1 domain and the adjacent domains. In fact, MID1 E3 ligase activity is slightly enhanced. However, the P151L mutation disrupted the ability of MID1 to catalyze the poly-ubiquitination of alpha4, a novel regulator of PP2A. This observation is consistent with results observed with the other three structure-destabilizing B-box1 mutations in targeting alpha4 but not PP2A. Alpha4 is shown to bind and sequester the catalytic subunit of PP2A and protect it from MID1-mediated ubiquitination and as a result, an increase in alpha4 can contribute to an increase in PP2A, playing a greater role in midline development during embryogenesis.


Subject(s)
Cleft Palate/genetics , Cleft Palate/metabolism , Esophagus/abnormalities , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Hypertelorism/genetics , Hypertelorism/metabolism , Hypospadias/genetics , Hypospadias/metabolism , Microtubule Proteins/chemistry , Microtubule Proteins/metabolism , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Sequence , Cleft Palate/pathology , Esophagus/metabolism , Esophagus/pathology , Genetic Diseases, X-Linked/pathology , Humans , Hypertelorism/pathology , Hypospadias/pathology , Microtubule Proteins/genetics , Microtubule Proteins/ultrastructure , Models, Molecular , Nuclear Proteins/genetics , Nuclear Proteins/ultrastructure , Protein Domains , Protein Phosphatase 2/metabolism , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Substrate Specificity , Transcription Factors/genetics , Transcription Factors/ultrastructure , Ubiquitin-Protein Ligases , Ubiquitination
15.
FEBS J ; 283(16): 3089-102, 2016 08.
Article in English | MEDLINE | ID: mdl-27367845

ABSTRACT

UNLABELLED: The human MID1 protein is required for the proper development during embryogenesis. Mutations of MID1 are associated with X-linked Opitz G syndrome, characterized by midline anomalies. MID1 associates with the microtubules and functions as an ubiquitin E3 ligase, targeting protein phosphatase 2A for ubiquitin-mediated regulation. The mechanism of microtubule association is not known. Recently, a 60-amino acid region termed the C-terminal subgroup One Signature (COS) box/domain was identified at the C-terminal end of the coiled-coil (CC) domain that facilitates microtubule localization. Insertion of the MID1 COS domain at the C-terminal end of the CC domain of a nonmicrotubule-associated TRIM protein confers microtubule localization. Here, we report the solution structure of the COS domain of MID1. The domain adopts a helix-loop-helix structure in which the N- and C-terminal ends are in close proximity. Hydrophobic residues stabilizing the interaction of the two α-helices form a central hydrophobic core. The loop separating the α-helices is structured, with two of its hydrophobic residues making contact with the central core. On the outer surface, positively charged residues form a distinct basic patch near the termini that we postulate is important for microtubule binding. A model of the structure of the preceding coiled-coil and COS domains (CC-COS) show that the COS domain forms a helical bundle at the C-terminal end of the CC domain similar to the spectrin-like fold observed with some known microtubule-binding proteins. Interestingly, the CC-COS domains bind to microtubules, demonstrating for the first time that MID1 can directly associate with the microtubules. DATABASE: Structural data are available in PDB database under the accession number 5IM8.


Subject(s)
Microtubule Proteins/chemistry , Microtubules/metabolism , Nuclear Proteins/chemistry , Transcription Factors/chemistry , Humans , Microtubule Proteins/metabolism , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/metabolism , Protein Conformation, alpha-Helical , Protein Domains , Transcription Factors/metabolism , Ubiquitin-Protein Ligases
16.
Curr Protoc Protein Sci ; 84: 6.13.1-6.13.24, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27038270

ABSTRACT

This unit describes a straightforward and efficient method of using sarkosyl to solubilize and recover difficult recombinant proteins, such as GST- and His6 -tagged fusion proteins, that are overexpressed in E. coli. This protocol is especially useful for rescuing recombinant proteins overexpressed in M9 minimal medium. Sarkosyl added to lysis buffers helps with both protein solubility and cell lysis. Higher percentage sarkosyl (up to 10%) can extract >95% of soluble protein from inclusion bodies. In the case of sarkosyl-solubilized GST-fusion proteins, batch-mode affinity purification requires addition of a specific ratio of Triton X-100 and CHAPS, while sarkosyl-solubilized His6 -tagged fusion proteins can be directly purified on Ni(2+) resin columns. Proteins purified by this method could be widely used in biological assays, structure analysis and mass spectrum assay.


Subject(s)
Cholic Acids/chemistry , Inclusion Bodies/chemistry , Octoxynol/chemistry , Recombinant Fusion Proteins/isolation & purification , Sarcosine/analogs & derivatives , Betaine/chemistry , Escherichia coli/genetics , Glutathione Transferase/genetics , Histidine/genetics , Protein Engineering/methods , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sarcosine/chemistry , Sarcosine/isolation & purification , Solubility , Sorbitol/chemistry
17.
PLoS One ; 9(9): e107537, 2014.
Article in English | MEDLINE | ID: mdl-25216264

ABSTRACT

MID1 catalyzes the ubiquitination of the protein alpha4 and the catalytic subunit of protein phosphatase 2A. Mutations within the MID1 Bbox1 domain are associated with X-linked Opitz G syndrome (XLOS). Our functional assays have shown that mutations of Ala130 to Val or Thr, Cys142 to Ser and Cys145 to Thr completely disrupt the polyubiquitination of alpha4. Using NMR spectroscopy, we characterize the effect of these mutations on the tertiary structure of the Bbox1 domain by itself and in tandem with the Bbox2 domain. The mutation of either Cys142 or Cys145, each of which is involved in coordinating one of the two zinc ions, results in the collapse of signal dispersion in the HSQC spectrum of the Bbox1 domain indicating that the mutant protein structure is unfolded. Each mutation caused the coordination of both zinc ions, which are ∼ 13 Šapart, to be lost. Although Ala130 is not involved in the coordination of a zinc ion, the Ala130Thr mutant Bbox1 domain yields a poorly dispersed HSQC spectrum similar to those of the Cys142Ser and Cys145Thr mutants. Interestingly, neither cysteine mutation affects the structure of the adjacent Bbox2 domain when the two Bbox domains are engineered in their native tandem Bbox1-Bbox2 protein construct. Dynamic light scattering measurements suggest that the mutant Bbox1 domain has an increased propensity to form aggregates compared to the wild type Bbox1 domain. These studies provide insight into the mechanism by which mutations observed in XLOS affect the structure and function of the MID1 Bbox1 domain.


Subject(s)
Cleft Palate/genetics , Esophagus/abnormalities , Genetic Diseases, X-Linked/genetics , Hypertelorism/genetics , Hypospadias/genetics , Microtubule Proteins/chemistry , Mutation , Nuclear Proteins/chemistry , Protein Phosphatase 2/genetics , Transcription Factors/chemistry , Amino Acid Sequence , Esophagus/chemistry , Humans , Male , Microtubule Proteins/genetics , Models, Molecular , Nuclear Proteins/genetics , Protein Phosphatase 2/chemistry , Protein Structure, Tertiary , Transcription Factors/genetics , Ubiquitin-Protein Ligases , Ubiquitination/genetics , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...