Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 6947, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37935654

ABSTRACT

Disease-causing mutations in genes encoding transcription factors (TFs) can affect TF interactions with their cognate DNA-binding motifs. Whether and how TF mutations impact upon the binding to TF composite elements (CE) and the interaction with other TFs is unclear. Here, we report a distinct mechanism of TF alteration in human lymphomas with perturbed B cell identity, in particular classic Hodgkin lymphoma. It is caused by a recurrent somatic missense mutation c.295 T > C (p.Cys99Arg; p.C99R) targeting the center of the DNA-binding domain of Interferon Regulatory Factor 4 (IRF4), a key TF in immune cells. IRF4-C99R fundamentally alters IRF4 DNA-binding, with loss-of-binding to canonical IRF motifs and neomorphic gain-of-binding to canonical and non-canonical IRF CEs. IRF4-C99R thoroughly modifies IRF4 function by blocking IRF4-dependent plasma cell induction, and up-regulates disease-specific genes in a non-canonical Activator Protein-1 (AP-1)-IRF-CE (AICE)-dependent manner. Our data explain how a single mutation causes a complex switch of TF specificity and gene regulation and open the perspective to specifically block the neomorphic DNA-binding activities of a mutant TF.


Subject(s)
Interferon Regulatory Factors , Lymphoma , Humans , B-Lymphocytes/metabolism , DNA , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Lymphoma/genetics
2.
Commun Biol ; 6(1): 4, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36596888

ABSTRACT

Hypertrophic cardiomyopathy is an inherited disorder due to mutations in contractile proteins that results in a stiff, hypercontractile myocardium. To understand the role of cardiac stiffness in disease progression, here we create an in vitro model of hypertrophic cardiomyopathy utilizing hydrogel technology. Culturing wild-type cardiac myocytes on hydrogels with a Young's Moduli (stiffness) mimicking hypertrophic cardiomyopathy myocardium is sufficient to induce a hypermetabolic mitochondrial state versus myocytes plated on hydrogels simulating healthy myocardium. Significantly, these data mirror that of myocytes isolated from a murine model of human hypertrophic cardiomyopathy (cTnI-G203S). Conversely, cTnI-G203S myocyte mitochondrial function is completely restored when plated on hydrogels mimicking healthy myocardium. We identify a mechanosensing feedback mechanism between the extracellular matrix and cytoskeletal network that regulates mitochondrial function under healthy conditions, but participates in the progression of hypertrophic cardiomyopathy pathophysiology resulting from sarcomeric gene mutations. Importantly, we pinpoint key 'linker' sites in this schema that may represent potential therapeutic targets.


Subject(s)
Cardiomyopathy, Hypertrophic , Mice , Humans , Animals , Feedback , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cytoskeleton/metabolism , Myocytes, Cardiac/metabolism , Troponin I/genetics , Troponin I/metabolism , Extracellular Matrix/metabolism , Hydrogels
3.
Protein Sci ; 31(5): e4311, 2022 05.
Article in English | MEDLINE | ID: mdl-35481653

ABSTRACT

Excitation-contraction coupling (ECC) is the physiological process in which an electrical signal originating from the central nervous system is converted into muscle contraction. In skeletal muscle tissue, the key step in the molecular mechanism of ECC initiated by the muscle action potential is the cooperation between two Ca2+ channels, dihydropyridine receptor (DHPR; voltage-dependent L-type calcium channel) and ryanodine receptor 1 (RyR1). These two channels were originally postulated to communicate with each other via direct mechanical interactions; however, the molecular details of this cooperation have remained ambiguous. Recently, it has been proposed that one or more supporting proteins are in fact required for communication of DHPR with RyR1 during the ECC process. One such protein that is increasingly believed to play a role in this interaction is the SH3 and cysteine-rich domain-containing protein 3 (STAC3), which has been proposed to bind a cytosolic portion of the DHPR α1S subunit known as the II-III loop. In this work, we present direct evidence for an interaction between a small peptide sequence of the II-III loop and several residues within the SH3 domains of STAC3 as well as the neuronal isoform STAC2. Differences in this interaction between STAC3 and STAC2 suggest that STAC3 possesses distinct biophysical features that are potentially important for its physiological interactions with the II-III loop. Therefore, this work demonstrates an isoform-specific interaction between STAC3 and the II-III loop of DHPR and provides novel insights into a putative molecular mechanism behind this association in the skeletal muscle ECC process.


Subject(s)
Calcium Channels, L-Type , Ryanodine Receptor Calcium Release Channel , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Excitation Contraction Coupling/physiology , Muscle, Skeletal/physiology , Protein Isoforms/metabolism
4.
Protein Sci ; 31(4): 787-796, 2022 04.
Article in English | MEDLINE | ID: mdl-34913532

ABSTRACT

Interferon regulatory factor 4 (IRF4) is an essential regulator in the development of many immune cells, including B- and T-cells and has been implicated directly in numerous hematological malignancies, including adult T-cell leukemia/lymphoma (ATLL). Recently, an activating mutation in the DNA-binding domain of IRF4 (IRF4K59R ) was found as a recurrent somatic mutation in ATLL patients. However, it remains unknown how this mutation gives rise to the observed oncogenic effect. To understand the mode of IRF4K59R -mediated gain of function in ATLL pathogenesis, we have determined the structural and affinity basis of IRF4K59R /DNA homodimer complex using X-ray crystallography and surface plasmon resonance. Our study shows that arginine substitution (R59) results in the reorientation of the side chain, enabling the guanidium group to interact with the phosphate backbone of the DNA helix. This markedly contrasts with the IRF4WT wherein the K59 interacts exclusively with DNA bases. Further, the arginine mutation causes enhanced DNA bending, enabling the IRF4K59R to interact more robustly with known DNA targets, as evidenced by increased binding affinity of the protein-DNA complex. Together, we demonstrate how key structural features underpin the basis for this activating mutation, thereby providing a molecular rationale for IRF4K59R -mediated ATLL development.


Subject(s)
Interferon Regulatory Factors , Leukemia-Lymphoma, Adult T-Cell , Adult , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Mutation
5.
Cell Calcium ; 96: 102403, 2021 06.
Article in English | MEDLINE | ID: mdl-33813182

ABSTRACT

The phosphoprotein AHNAK is a large, ubiquitously expressed scaffolding protein involved in mediating a host of protein-protein interactions. This enables AHNAK to participate in various multi-protein complexes thereby orchestrating a range of diverse biological processes, including tumour suppression, immune regulation and cell architecture maintenance. A less studied but nonetheless equally important function occurs in calcium homeostasis. It does so by largely interacting with the L-type voltage-gated calcium channel (LVGCC) present in the plasma membrane of excitable cells such as muscles and neurons. Several studies have characterized the underlying basis of AHNAK's functional role in calcium channel modulation, which has led to a greater understanding of this cellular process and its associated pathologies. In this article we review and examine recent advances relating to the physiological aspects of AHNAK in calcium regulation. Specifically, we will provide a broad overview of AHNAK including its structural makeup and its interaction with several isoforms of LVGCC, and how these molecular interactions regulate calcium modulation across various tissues and their implication in muscle and neuronal function.


Subject(s)
Calcium/metabolism , Homeostasis/physiology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Animals , Binding Sites/physiology , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Humans
6.
Nucleic Acids Res ; 49(4): 2255-2265, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33533913

ABSTRACT

Interferon regulatory factor 4 (IRF4) is a key transcription factor (TF) in the regulation of immune cells, including B and T cells. It acts by binding DNA as both a homodimer and, in conjunction with other TFs, as a heterodimer. The choice of homo and heterodimeric/ DNA interactions is a critical aspect in the control of the transcriptional program and cell fate outcome. To characterize the nature of this interaction in the homodimeric complex, we have determined the crystal structure of the IRF4/ISRE homodimeric complex. We show that the complex formation is aided by a substantial DNA deformation with co-operative binding achieved exclusively through protein-DNA contact. This markedly contrasts with the heterodimeric form where DNA bound IRF4 is shown to physically interact with PU.1 TF to engage EICE1. We also show that the hotspot residues (Arg98, Cys99 and Asn102) contact both consensus and non-consensus sequences with the L1 loop exhibiting marked flexibility. Additionally, we identified that IRF4L116R, a mutant associated with chronic lymphocytic leukemia, binds more robustly to DNA thereby providing a rationale for the observed gain of function. Together, we demonstrate key structural differences between IRF4 homo and heterodimeric complexes, thereby providing molecular insights into IRF4-mediated transcriptional regulation.


Subject(s)
DNA/chemistry , Interferon Regulatory Factors/chemistry , DNA/metabolism , Dimerization , Gain of Function Mutation , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Models, Molecular , Protein Binding , Protein Domains , Protein Multimerization , Proto-Oncogene Proteins/chemistry , Trans-Activators/chemistry
7.
Biophys Rev ; 13(6): 1219-1227, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35059038

ABSTRACT

Interferon regulatory factor 4 (IRF4) is a lymphoid transcription factor and a key regulator in the development of various immune cells, including T and B cells. It is well-known that IRF4 controls numerous decision-making processes relating to B cell development, including differentiation, maturation, and signalling. Consequently, genetic alterations that affect the functional aspects of IRF4 can result in clonal transformation and have been identified in various lymphoid malignancies. Over the last decades, a series of studies have demonstrated the critical cellular and structural basis underpinning IRF4-mediated B cell development and associated malignancies. In this review, we will briefly summarise the recent advances in understanding IRF4-mediated B cell development and related malignancies, with a particular focus on the molecular aspects that govern these processes.

8.
Nat Commun ; 10(1): 5242, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748533

ABSTRACT

Type I and type II natural killer T (NKT) cells are restricted to the lipid antigen-presenting molecule CD1d. While we have an understanding of the antigen reactivity and function of type I NKT cells, our knowledge of type II NKT cells in health and disease remains unclear. Here we describe a population of type II NKT cells that recognise and respond to the microbial antigen, α-glucuronosyl-diacylglycerol (α-GlcADAG) presented by CD1d, but not the prototypical type I NKT cell agonist, α-galactosylceramide. Surprisingly, the crystal structure of a type II NKT TCR-CD1d-α-GlcADAG complex reveals a CD1d F'-pocket-docking mode that contrasts sharply with the previously determined A'-roof positioning of a sulfatide-reactive type II NKT TCR. Our data also suggest that diverse type II NKT TCRs directed against distinct microbial or mammalian lipid antigens adopt multiple recognition strategies on CD1d, thereby maximising the potential for type II NKT cells to detect different lipid antigens.


Subject(s)
Antigens, CD1d/immunology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen Presentation , Antigens, CD1d/metabolism , Crystallography, X-Ray , Flow Cytometry , Galactosylceramides/immunology , Glycolipids/immunology , Mice , Mice, Knockout , Molecular Docking Simulation , Natural Killer T-Cells/metabolism , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/metabolism
9.
Cell Rep ; 29(1): 151-161.e5, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31577945

ABSTRACT

The NLRP3 inflammasome is a cytosolic complex sensing phagocytosed material and various damage-associated molecular patterns, triggering production of the pro-inflammatory cytokines interleukin-1 beta (IL)-1ß and IL-18 and promoting pyroptosis. Here, we characterize glutathione transferase omega 1-1 (GSTO1-1), a constitutive deglutathionylating enzyme, as a regulator of the NLRP3 inflammasome. Using a small molecule inhibitor of GSTO1-1 termed C1-27, endogenous GSTO1-1 knockdown, and GSTO1-1-/- mice, we report that GSTO1-1 is involved in NLRP3 inflammasome activation. Mechanistically, GSTO1-1 deglutathionylates cysteine 253 in NIMA related kinase 7 (NEK7) to promote NLRP3 activation. We therefore identify GSTO1-1 as an NLRP3 inflammasome regulator, which has potential as a drug target to limit NLRP3-mediated inflammation.


Subject(s)
Glutathione Transferase/metabolism , Inflammasomes/metabolism , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Cytokines/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL
11.
Cell Chem Biol ; 25(5): 571-584.e8, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29576533

ABSTRACT

Glycosylceramides that activate CD1d-restricted invariant natural killer T (iNKT) cells have potential therapeutic applications for augmenting immune responses against cancer and infections. Previous studies using mouse models identified sphinganine variants of α-galactosylceramide as promising iNKT cell activators that stimulate cytokine responses with a strongly proinflammatory bias. However, the activities of sphinganine variants in mice have generally not translated well to studies of human iNKT cell responses. Here, we show that strongly proinflammatory and anti-tumor iNKT cell responses were achieved in mice by a variant of α-galactosylceramide that combines a sphinganine base with a hydrocinnamoyl ester on C6″ of the sugar. Importantly, the activities observed with this variant were largely preserved for human iNKT cell responses. Structural and in silico modeling studies provided a mechanistic basis for these findings and suggested basic principles for capturing useful properties of sphinganine analogs of synthetic iNKT cell activators in the design of immunotherapeutic agents.


Subject(s)
Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , Galactosylceramides/chemistry , Galactosylceramides/pharmacology , Lymphocyte Activation/drug effects , Natural Killer T-Cells/drug effects , Neoplasms/therapy , Adolescent , Adult , Aged , Animals , Antigens, CD1d/immunology , Cell Line, Tumor , Cells, Cultured , Female , Humans , Immunotherapy , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Natural Killer T-Cells/immunology , Neoplasms/immunology
12.
Biomol NMR Assign ; 12(2): 253-257, 2018 10.
Article in English | MEDLINE | ID: mdl-29594929

ABSTRACT

Ahnak is a ~ 700 kDa polypeptide that was originally identified as a tumour-related nuclear phosphoprotein, but later recognized to play a variety of diverse physiological roles related to cell architecture and migration. A critical function of Ahnak is modulation of Ca2+ signaling in cardiomyocytes by interacting with the ß subunit of the L-type Ca2+ channel (CaV1.2). Previous studies have identified the C-terminal region of Ahnak, designated as P3 and P4 domains, as a key mediator of its functional activity. We report here the nearly complete 1H, 13C and 15N backbone NMR chemical shift assignments of the 11 kDa C-terminal P4 domain of Ahnak. This study lays the foundations for future investigations of functional dynamics, structure determination and interaction site mapping of the CaV1.2-Ahnak complex.


Subject(s)
Membrane Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Binding Sites , Calcium Channels, L-Type/metabolism , Membrane Proteins/metabolism , Protein Domains , Protein Subunits/chemistry , Protein Subunits/metabolism
13.
Proc Natl Acad Sci U S A ; 115(6): E1204-E1213, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29351991

ABSTRACT

MHC class I-like CD1 molecules have evolved to present lipid-based antigens to T cells. Differences in the antigen-binding clefts of the CD1 family members determine the conformation and size of the lipids that are presented, although the factors that shape CD1 diversity remain unclear. In mice, two homologous genes, CD1D1 and CD1D2, encode the CD1d protein, which is essential to the development and function of natural killer T (NKT) cells. However, it remains unclear whether both CD1d isoforms are equivalent in their antigen presentation capacity and functions. Here, we report that CD1d2 molecules are expressed in the thymus of some mouse strains, where they select functional type I NKT cells. Intriguingly, the T cell antigen receptor repertoire and phenotype of CD1d2-selected type I NKT cells in CD1D1-/- mice differed from CD1d1-selected type I NKT cells. The structures of CD1d2 in complex with endogenous lipids and a truncated acyl-chain analog of α-galactosylceramide revealed that its A'-pocket was restricted in size compared with CD1d1. Accordingly, CD1d2 molecules could not present glycolipid antigens with long acyl chains efficiently, favoring the presentation of short acyl chain antigens. These results indicate that the two CD1d molecules present different sets of self-antigen(s) in the mouse thymus, thereby impacting the development of invariant NKT cells.


Subject(s)
Antigen Presentation/immunology , Antigens, CD1d/physiology , Cell Differentiation , Glycolipids/immunology , Killer Cells, Natural/immunology , Thymus Gland/immunology , Animals , Cells, Cultured , Crystallography, X-Ray , Killer Cells, Natural/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Conformation , Protein Isoforms , Thymus Gland/cytology
14.
PLoS One ; 9(4): e93008, 2014.
Article in English | MEDLINE | ID: mdl-24699522

ABSTRACT

Cysteine proteases play a crucial role in the development of the human malaria parasites Plasmodium falciparum and Plasmodium vivax. Our earlier studies demonstrated that these enzymes are equipped with specific domains for defined functions and further suggested the mechanism of activation of cysteine proteases. The activities of these proteases are regulated by a new class of endogenous inhibitors of cysteine proteases (ICPs). Structural studies of the ICPs of Trypanosoma cruzi (chagasin) and Plasmodium berghei (PbICP) indicated that three loops (termed BC, DE, and FG) are crucial for binding to target proteases. Falstatin, an ICP of P. falciparum, appears to play a crucial role in invasion of erythrocytes and hepatocytes. However, the mechanism of inhibition of cysteine proteases by falstatin has not been established. Our study suggests that falstatin is the first known ICP to function as a multimeric protein. Using site-directed mutagenesis, hemoglobin hydrolysis assays and peptide inhibition studies, we demonstrate that the BC loop, but not the DE or FG loops, inhibits cysteine proteases of P. falciparum and P. vivax via hydrogen bonds. These results suggest that the BC loop of falstatin acts as a hot-spot target for inhibiting malarial cysteine proteases. This finding suggests new strategies for the development of anti-malarial agents based on protease-inhibitor interactions.


Subject(s)
Cysteine Proteases/metabolism , Cysteine Proteinase Inhibitors/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/parasitology , Plasmodium falciparum/enzymology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Amino Acid Sequence , Cross Reactions , Cysteine Proteases/chemistry , Erythrocytes/pathology , Hemoglobins/metabolism , Humans , Hydrogen Bonding , Hydrolysis , Malaria, Falciparum/pathology , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Plasmodium falciparum/genetics , Protein Conformation , Protein Multimerization , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sequence Homology, Amino Acid
15.
PLoS One ; 7(10): e47227, 2012.
Article in English | MEDLINE | ID: mdl-23077573

ABSTRACT

The Plasmodium falciparum cysteine proteases falcipain-2 and falcipain-3 are major hemoglobinases and potential antimalarial drug targets. Our previous studies demonstrated that these enzymes are equipped with specific domains for specific functions. Structural and functional analysis of falcipains showed that they have unique domains including a refolding domain and a hemoglobin binding domain. As with many proteases, falcipain-2 and falcipain-3 are synthesized as inactive zymogens. However, it is not known how these enzymes get activated for hemoglobin hydrolysis. In this study, we are presenting the first evidence that salt bridges and hydrophobic interactions are required for the auto activation of cysteine proteases of P.falciparum. To investigate the mechanism of activation of these enzymes, we expressed the wild type protein as well as different mutants in E.coli. Refolding was assessed by circular dichroism. Both CD and trans activation data showed that the wild type enzymes and mutants are rich in secondary structures with similar folds. Our study revealed that prodomain-mature domain of falcipain-2 and falcipain-3 interacts via salt bridges and hydrophobic interactions. We mutated specific residues of falcipain-2 and falcipain-3, and evaluated their ability to undergo auto processing. Mutagenesis result showed that two salt bridges (Arg¹85- Glu²²¹, Glu²¹°- Lys4°³) in falcipain-2, and one salt bridge (Arg²°²-Glu²³8) in falcipain-3, play crucial roles in the activation of these enzymes. Further study revealed that hydrophobic interactions present both in falcipain-2 (Phe²¹4 Trp449 Trp45³) and falcipain-3 (Phe²³¹ Trp457 Trp46¹) also play important roles in the activation of these enzymes. Our results revealed the interactions involved in auto processing of two major hemoglobinases of malaria parasite.


Subject(s)
Cysteine Endopeptidases/genetics , Enzyme Activation/genetics , Malaria, Falciparum/enzymology , Plasmodium falciparum/enzymology , Amino Acid Sequence , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Hemoglobins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Malaria, Falciparum/parasitology , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...