Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cancer Sci ; 112(9): 3419-3426, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34159709

ABSTRACT

Acute myeloid leukemia (AML) is hierarchically organized by self-renewing leukemic stem cells (LSCs). LSCs originate from hematopoietic stem cells (HSCs) by acquiring multistep leukemogenic events. To specifically eradicate LSCs, while keeping normal HSCs intact, the discrimination of LSCs from HSCs is important. We have identified T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) as an LSC-specific surface molecule in human myeloid malignancies and demonstrated its essential function in maintaining the self-renewal ability of LSCs. TIM-3 has been intensively investigated as a "coinhibitory" or "immune checkpoint" molecule of T cells. However, little is known about its distinct function in T cells and myeloid malignancies. In this review, we discuss the structure of TIM-3 and its function in normal blood cells and LSCs, emphasizing the specific signaling pathways involved, as well as the therapeutic applications of TIM-3 molecules in human myeloid malignancies.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/metabolism , Hepatitis A Virus Cellular Receptor 2/chemistry , Hepatitis A Virus Cellular Receptor 2/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Animals , Humans , Leukemia, Myeloid, Acute/pathology , Molecular Structure , T-Lymphocytes/metabolism
2.
Microb Pathog ; 144: 104198, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32283259

ABSTRACT

HTLV-1, the first human oncogenic retrovirus, is a type C retrovirus that belongs to the Deltaretrovirus genus. The HTLV-1 genome has 8.5 kbp length, and consists of major genes such as gag, pol, pro, env, and pX region. This retrovirus is considered as one of the most deadly infectious agent for peripheral-blood mononuclear cells (PBMC). The infection of HTLV-1 can lead to dangerous complications, such as infective dermatitis (ID), uveitis, arthritis, lymphadenitis, arthropathies, Sjögren's Syndrome (SS), and particularly HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) or Adult T-Cell Leukemia Lymphoma (ATLL). At the moment, Zidovudine (AZT) plus IFN-α is the only treatment available for HTLV-1 infections. Based on scientific studies, alongside the therapeutic regimens, intrinsic mechanisms also play a determinant role in reducing the signs of disease. Programmed cell death-1 (PD-1) signaling pathway, one of the most important checkpoints, has recently received interest, such as the development of a novel generation of anti-tumors. In the present study, we discuss the role of PD-1 signaling pathway in HTLV-1 infection as well as its application as a novel approach for treatment of HTLV-1 infections.


Subject(s)
HTLV-I Infections/drug therapy , Human T-lymphotropic virus 1/drug effects , Human T-lymphotropic virus 1/genetics , Molecular Targeted Therapy/methods , Programmed Cell Death 1 Receptor/drug effects , Signal Transduction/drug effects , Adult , Antigens, CD/chemistry , CTLA-4 Antigen/chemistry , Chronic Disease , HTLV-I Infections/complications , HTLV-I Infections/virology , Hepatitis A Virus Cellular Receptor 2/chemistry , Humans , Interferon-alpha/pharmacology , Leukocytes, Mononuclear/virology , Paraparesis, Tropical Spastic , Programmed Cell Death 1 Receptor/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Receptors, Immunologic/chemistry , Zidovudine/pharmacology , Lymphocyte Activation Gene 3 Protein
3.
Science ; 368(6486): 78-84, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32241946

ABSTRACT

The design of modular protein logic for regulating protein function at the posttranscriptional level is a challenge for synthetic biology. Here, we describe the design of two-input AND, OR, NAND, NOR, XNOR, and NOT gates built from de novo-designed proteins. These gates regulate the association of arbitrary protein units ranging from split enzymes to transcriptional machinery in vitro, in yeast and in primary human T cells, where they control the expression of the TIM3 gene related to T cell exhaustion. Designed binding interaction cooperativity, confirmed by native mass spectrometry, makes the gates largely insensitive to stoichiometric imbalances in the inputs, and the modularity of the approach enables ready extension to three-input OR, AND, and disjunctive normal form gates. The modularity and cooperativity of the control elements, coupled with the ability to de novo design an essentially unlimited number of protein components, should enable the design of sophisticated posttranslational control logic over a wide range of biological functions.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/chemistry , Protein Engineering , Protein Interaction Maps , Protein Processing, Post-Translational , Hepatitis A Virus Cellular Receptor 2/genetics , Humans , Logic , Mass Spectrometry , Synthetic Biology , T-Lymphocytes/metabolism , Transcription, Genetic , Yeasts/metabolism
4.
Semin Immunol ; 42: 101302, 2019 04.
Article in English | MEDLINE | ID: mdl-31604535

ABSTRACT

T cell inhibitory co-receptors play a crucial role in maintaining the balance between physiologic immune responses and maladaptive ones. T cell immunoglobulin and mucin domain-containing-3 (Tim-3) is a unique inhibitory co-receptor in that its expression is chiefly restricted to interferon (IFN)γ-producing CD4+ and CD8+ T cells. Early reports firmly established its importance in maintaining peripheral tolerance in transplantation and autoimmunity. However, it has become increasingly clear that Tim-3 expression on T cells, together with other check-point molecules, in chronic infections and cancers can hinder productive immune responses. In this review, we outline what is currently known about the regulation of Tim-3 expression, its ligands and signaling. We discuss both its salutary and deleterious function in immune disorders, as well as the T cell-extrinsic and -intrinsic factors that regulate its function.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/immunology , Immune Tolerance , T-Lymphocytes/immunology , Animals , Autoimmunity , Hepatitis A Virus Cellular Receptor 2/chemistry , Humans , Infections/immunology , Ligands , Neoplasms/immunology , Protein Conformation
5.
Sci Rep ; 8(1): 17512, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504845

ABSTRACT

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


Subject(s)
Crystallography, X-Ray/methods , Hepatitis A Virus Cellular Receptor 2/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Humans , Mice , Protein Conformation , Sequence Homology, Amino Acid
6.
Planta Med ; 84(16): 1201-1212, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29742762

ABSTRACT

Native mass spectrometry detection of ligand-protein complexes allowed rapid detection of natural product binders of apo and calcium-bound S100A4 (a member of the metal binding protein S100 family), T cell/transmembrane, immunoglobulin (Ig), and mucin protein 3, and T cell immunoreceptor with Ig and ITIM (immunoreceptor tyrosine-based inhibitory motif) domains precursor protein from extracts and fractions. Based on molecular weight common hits were detected binding to all four proteins. Seven common hits were identified as apigenin 6-C-ß-D-glucoside 8-C-α-L-arabinoside, sweroside, 4',5-dihydroxy-7-methoxyflavanone-6-C-rutinoside, loganin acid, 6-C-glucosylnaringenin, biochanin A 7-O-rutinoside and quercetin 3-O-rutinoside. Mass guided isolation and NMR identification of hits confirmed the mass accuracy of the ligand in the ligand-protein MS complexes. Thus, molecular weight ID from ligand-protein complexes by electrospray ionization Fourier transform mass spectrometry allowed rapid dereplication. Native mass spectrometry using electrospray ionization Fourier transform mass spectrometry is a tool for dereplication and metabolomics analysis.


Subject(s)
Drug Evaluation, Preclinical/methods , Hepatitis A Virus Cellular Receptor 2/metabolism , Receptors, Immunologic/metabolism , S100 Calcium-Binding Protein A4/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Calcium/chemistry , Calcium/metabolism , Fourier Analysis , Hepatitis A Virus Cellular Receptor 2/analysis , Hepatitis A Virus Cellular Receptor 2/chemistry , Magnetic Resonance Spectroscopy , Molecular Weight , Plant Extracts/analysis , Plant Extracts/metabolism , Receptors, Immunologic/analysis , Receptors, Immunologic/chemistry , S100 Calcium-Binding Protein A4/analysis , S100 Calcium-Binding Protein A4/chemistry
7.
Immunology ; 153(1): 71-83, 2018 01.
Article in English | MEDLINE | ID: mdl-28799242

ABSTRACT

The Nod-like receptor protein 3 (NLRP3) inflammasome plays roles in host defence against invading pathogens and in the development of autoimmune damage. Strict regulation of these responses is important to avoid detrimental effects. Here, we demonstrate that T cell Ig mucin-3 (Tim-3), an immune checkpoint inhibitor, inhibits NLRP3 inflammasome activation by damping basal and lipopolysaccharide-induced nuclear factor-κB-mediated up-regulation of NLRP3 and interleukin-1ß during the priming step and basal and ATP/lipopolysaccharide-induced ATP production, K+ efflux, and reactive oxygen species production during the activation step. Residues Y256/Y263 in the C-terminal region of Tim-3 are required for these inhibitory effects on the NLRP3 inflammasome. In mice with alum-induced peritonitis, blockade of Tim-3 exacerbates peritonitis by overcoming the inhibitory effect of Tim-3 on NLRP3 inflammasome activation, while transgenic expression of Tim-3 attenuates inflammation by inhibiting NLRP3 inflammasome activation. Our results show that Tim-3 is a critical negative regulator of NLRP3 inflammasome and provides a potential target for intervention of diseases with uncontrolled inflammasome activation.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peritonitis/immunology , Peritonitis/metabolism , Adenosine Triphosphate/biosynthesis , Adult , Animals , Case-Control Studies , Caspase 1 , Cell Line , Disease Models, Animal , Female , Hepatitis A Virus Cellular Receptor 2/chemistry , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Middle Aged , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Peritonitis/pathology , Potassium/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Reactive Oxygen Species/metabolism , Signal Transduction , Young Adult
8.
Sci Rep ; 7(1): 13579, 2017 10 19.
Article in English | MEDLINE | ID: mdl-29051586

ABSTRACT

In the face of chronic cancers and protracted viral infections, human immune cells are known to adopt an exhausted state in which their effector functions are lost. In recent years, a number of inhibitory receptors have been connected to the immune cell exhaustion phenotype; furthermore, ligands capable of activating these receptors have been discovered. The molecular mechanisms by which these ligands affect the exhausted states of immune cells, however, are largely unknown. Here, we present the results of molecular dynamics simulations of one potential exhaustion-associated system: the complex of human inhibitory receptor TIM3 (hTIM3) and its ligand phosphatidylserine (PSF). We find that PSF fundamentally alters the electrostatic environment within hTIM3's Ca2+ binding site, facilitating the formation of a salt bridge and freeing a tyrosine-containing strand. This liberated tyrosine then collapses into a nearby hydrophobic pocket, anchoring a modified conformational ensemble typified by a ß-strand rearrangement. The "electrostatic switching/hydrophobic anchoring" mechanism of conformational modulation reported here suggests a new type of process by which TIM3 activation might be achieved. This work also highlights strategies by which PSF-mediated conformational change could be controlled, either through administration of small molecules, execution of mutations, or modification of receptor phosphorylation states.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/chemistry , Hepatitis A Virus Cellular Receptor 2/metabolism , Phosphatidylserines/chemistry , Binding Sites , Immunoglobulin Domains , Models, Molecular , Molecular Dynamics Simulation , Phosphatidylserines/metabolism , Phosphorylation , Protein Conformation
9.
Mol Ther ; 25(10): 2280-2288, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28800954

ABSTRACT

T cell immunoglobulin-3 (TIM-3) is a negative regulator of interferon-γ (IFN-γ) secreting CD4+ T cells and CD8+ T cytotoxic cells. Recent studies have highlighted the role of TIM-3 as an important mediator of CD8+ T cell exhaustion in the setting of chronic viral infections and cancer. In murine tumor models, antibody blockade of TIM-3 with anti-TIM-3 antibodies as monotherapy has no or minimal antitumor activity, suggesting that TIM-3 signaling exerts an accessory or amplifying effect in keeping immune responses in check. Using a combined bead and cell-based systemic evolution of ligands by exponential enrichment (SELEX) protocol, we have isolated nuclease-resistant oligonucleotide aptamer ligands that bind to cell-associated TIM-3 with high affinity and specificity. A trimeric form of the TIM-3 aptamer blocked the interaction of TIM-3 with Galectin-9, reduced cell death, and enhanced survival, proliferation, and cytokine secretion in vitro. In tumor-bearing mice, the aptamer delayed tumor growth as monotherapy and synergized with PD-1 antibody in prolonging the survival of the tumor-bearing mice. Both in vitro and in vivo, the trimeric aptamer displayed superior activity compared to the currently used RMT3-23 monoclonal antibody. This study suggests that multi-valent aptamers could represent an alternative platform to generate potent ligands to manipulate the function of TIM-3 and other immune modulatory receptors.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Hepatitis A Virus Cellular Receptor 2/chemistry , Immunotherapy/methods , Animals , Aptamers, Nucleotide/chemistry , CHO Cells , Cricetulus , Female , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes , T-Lymphocytes, Cytotoxic/metabolism
10.
J Leukoc Biol ; 102(6): 1313-1322, 2017 12.
Article in English | MEDLINE | ID: mdl-28754800

ABSTRACT

T cell Ig and mucin domain-containing molecule 3 (Tim-3) has been found to play important roles in autoimmune diseases, but whether Tim-3-mediated engulfment of apoptotic cells is involved in systemic lupus erythematosus (SLE) remains to be elucidated. In this study, we verified the role of human Tim-3 (hTim-3) as the receptor of phosphatidylserine (PS) in human embryonic kidney (HEK)293 cells, which initiated the engulfment of apoptotic cells. Both IgV and the mucin domain of Tim-3 were crucial in the phagocytosis of apoptotic cells, and there existed the key cytoplasmic domain for signal transduction. Alanine at 111, locating around the FG-CC' loop of hTim-3, was necessary for its engulfment of apoptotic cells. In accordance, Tim-3 on CD14+ cells negatively correlated with the percentage of peripheral apoptotic cells in control subjects. However, although Tim-3 was significantly increased on CD14+ cells in SLE patients, peripheral apoptotic cells remained much higher than those in control subjects. Tim-3 on CD14+ cells showed positive correlation with percentage of apoptotic cells and level of dsDNA, indicating the involvement of Tim-3 in SLE. Accordingly, soluble Tim-3 (sTim-3) was significantly increased in plasma of SLE patients, which might contribute to higher expression of a disintegrin and metalloproteinase (ADAM)-10. Pretreatment with both plasma from SLE patients and recombinant sTim-3 greatly inhibited hTim-3-initiated phagocytosis of apoptotic cells. Furthermore, anti-Tim-3 antibody depletion of plasma from SLE patients reversed the decreased phagocytosis of apoptotic cells. Collectively, our data suggest that sTim-3 might play inhibitory roles in impaired Tim-3-mediated clearance of apoptotic cells in SLE.


Subject(s)
Apoptosis , Hepatitis A Virus Cellular Receptor 2/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Phagocytosis , Adult , Annexin A5/metabolism , Case-Control Studies , Cell Count , Cell Line , Female , Hepatitis A Virus Cellular Receptor 2/chemistry , Humans , Lipopolysaccharide Receptors/metabolism , Lupus Erythematosus, Systemic/blood , Male , Monocytes/metabolism , Protein Domains , Recombinant Proteins/pharmacology , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...