Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(5): 736-737, 2023 05.
Article in English | MEDLINE | ID: mdl-36997672
2.
Nature ; 618(7963): 159-168, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225977

ABSTRACT

Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.


Subject(s)
Nerve Regeneration , Humans , Neoplasms/drug therapy , Nerve Regeneration/drug effects , Protein Isoforms/agonists , Signal Transduction/drug effects , Class I Phosphatidylinositol 3-Kinases/chemistry , Class I Phosphatidylinositol 3-Kinases/drug effects , Cardiotonic Agents/pharmacology , Animals , Biocatalysis/drug effects , Protein Conformation/drug effects , Neurites/drug effects , Reperfusion Injury/prevention & control , Nerve Crush , Cell Proliferation/drug effects
3.
J Clin Immunol ; 40(7): 987-1000, 2020 10.
Article in English | MEDLINE | ID: mdl-32671674

ABSTRACT

Autoinflammatory diseases (AIDs) were first described as clinical disorders characterized by recurrent episodes of seemingly unprovoked sterile inflammation. In the past few years, the identification of novel AIDs expanded their phenotypes toward more complex clinical pictures associating vasculopathy, autoimmunity, or immunodeficiency. Herein, we describe two unrelated patients suffering since the neonatal period from a complex disease mainly characterized by severe sterile inflammation, recurrent bacterial infections, and marked humoral immunodeficiency. Whole-exome sequencing detected a novel, de novo heterozygous PLCG2 variant in each patient (p.Ala708Pro and p.Leu845_Leu848del). A clear enhanced PLCγ2 activity for both variants was demonstrated by both ex vivo calcium responses of the patient's B cells to IgM stimulation and in vitro assessment of PLC activity. These data supported the autoinflammation and PLCγ2-associated antibody deficiency and immune dysregulation (APLAID) diagnosis in both patients. Immunological evaluation revealed a severe decrease of immunoglobulins and B cells, especially class-switched memory B cells, with normal T and NK cell counts. Analysis of bone marrow of one patient revealed a reduced immature B cell fraction compared with controls. Additional investigations showed that both PLCG2 variants activate the NLRP3-inflammasome through the alternative pathway instead of the canonical pathway. Collectively, the evidences here shown expand APLAID diversity toward more severe phenotypes than previously reported including dominantly inherited agammaglobulinemia, add novel data about its genetic basis, and implicate the alternative NLRP3-inflammasome activation pathway in the basis of sterile inflammation.


Subject(s)
Agammaglobulinemia/diagnosis , Agammaglobulinemia/genetics , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/genetics , Mutation , Phospholipase C gamma/genetics , Adolescent , Agammaglobulinemia/therapy , Autoimmunity/genetics , Biomarkers , Caspase 1/metabolism , Child , Cytokines/metabolism , DNA Mutational Analysis , Female , Genetic Association Studies , Genetic Predisposition to Disease , Hereditary Autoinflammatory Diseases/therapy , Humans , Inflammasomes/metabolism , Male , Pedigree , Phenotype , Phospholipase C gamma/chemistry , Phospholipase C gamma/metabolism , Structure-Activity Relationship
4.
Int J Mol Sci ; 21(9)2020 May 01.
Article in English | MEDLINE | ID: mdl-32370101

ABSTRACT

Selective FGFR inhibitors such as infigratinib (BGJ398) and erdafitinib (JNJ-42756493) have been evaluated in clinical trials for cancers with FGFR3 molecular alterations, particularly in urothelial carcinoma patients. However, a substantial proportion of these patients (up to 50%) display intrinsic resistance to these drugs and receive minimal clinical benefit. There is thus an unmet need for alternative therapeutic strategies to overcome primary resistance to selective FGFR inhibitors. In this study, we demonstrate that cells expressing cancer-associated activating FGFR3 mutants and the FGFR3-TACC3 fusion showed primary resistance to infigratinib in long-term colony formation assays in both NIH-3T3 and urothelial carcinoma models. We find that expression of these FGFR3 molecular alterations resulted in elevated constitutive Src activation compared to wildtype FGFR3 and that cells co-opted this pathway as a means to achieve intrinsic resistance to infigratinib. Targeting the Src pathway with low doses of the kinase inhibitor dasatinib synergistically sensitized multiple urothelial carcinoma lines harbouring endogenous FGFR3 alterations to infigratinib. Our data provide preclinical rationale that supports the use of dasatinib in combination with selective FGFR inhibitors as a means to overcome intrinsic drug resistance in the salvage therapy setting in urothelial cancer patients with FGFR3 molecular alterations.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Signal Transduction/drug effects , Urologic Neoplasms/metabolism , src-Family Kinases/metabolism , Animals , Cell Line, Tumor , Humans , Mice , Microtubule-Associated Proteins/genetics , Molecular Targeted Therapy , NIH 3T3 Cells , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Kinase Inhibitors/therapeutic use , Urologic Neoplasms/drug therapy , Urologic Neoplasms/etiology , Urologic Neoplasms/pathology
5.
Trends Biochem Sci ; 39(12): 603-11, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25456276

ABSTRACT

The surge in genetic and genomic investigations over the past 5 years has resulted in many discoveries of causative variants relevant to disease pathophysiology. Although phospholipase C (PLC) enzymes have long been recognized as important components in intracellular signal transmission, it is only recently that this approach highlighted their role in disease development through gain-of-function mutations. In this review we describe the new findings that link the PLCγ family to immune disorders and cancer, and illustrate further efforts to elucidate the molecular mechanisms that underpin their dysfunction.


Subject(s)
Immune System Diseases/metabolism , Models, Biological , Models, Molecular , Neoplasms/metabolism , Phospholipase C gamma/metabolism , Second Messenger Systems , Animals , Diglycerides/metabolism , Humans , Immune System Diseases/immunology , Inositol 1,4,5-Trisphosphate/metabolism , Mutation , Neoplasms/genetics , Phospholipase C gamma/chemistry , Phospholipase C gamma/genetics , Protein Conformation
6.
Biophys J ; 115(1): 31-45, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29972810

ABSTRACT

Phosphatidylinositol phospholipase Cγ (PLCγ) is an intracellular membrane-associated second-messenger signaling protein activated by tyrosine kinases such as fibroblast growth factor receptor 1. PLCγ contains the regulatory γ-specific array (γSA) comprising a tandem Src homology 2 (SH2) pair, an SH3 domain, and a split pleckstrin homology domain. Binding of an activated growth factor receptor to γSA leads to Tyr783 phosphorylation and consequent PLCγ activation. Several disease-relevant mutations in γSA have been identified; all lead to elevated phospholipase activity. In this work, we describe an allosteric mechanism that connects the Tyr783 phosphorylation site to the nSH2-cSH2 junction and involves dynamic interactions between the cSH2-SH3 linker and cSH2. Molecular dynamics simulations of the tandem SH2 protein suggest that Tyr783 phosphorylation is communicated to the nSH2-cSH2 junction by modulating cSH2 binding to sections of the cSH2-SH3 linker. NMR chemical shift perturbation analyses for designed tandem SH2 constructs reveal combined fast and slow dynamic processes that can be attributed to allosteric communication involving these regions of the protein, establishing an example in which complex N-site exchange can be directly inferred from 1H,15N-HSQC spectra. Furthermore, in tandem SH2 and γSA constructs, molecular dynamics and NMR results show that the Arg687Trp mutant in PLCγ1 (equivalent to the cancer mutation Arg665Trp in PLCγ2) perturbs the dynamic allosteric pathway. This combined experimental and computational study reveals a rare example of multistate kinetics involved in a dynamic allosteric process that is modulated in the context of a disease-relevant mutation. The allosteric influences and the weakened binding of the cSH2-SH3 linker to cSH2 should be taken into account in any more holistic investigation of PLCγ regulation.


Subject(s)
Molecular Dynamics Simulation , Mutation , Neoplasms/genetics , Nuclear Magnetic Resonance, Biomolecular , Phospholipase C gamma/chemistry , Phospholipase C gamma/metabolism , Allosteric Regulation , Phospholipase C gamma/genetics , Phosphorylation , src Homology Domains
7.
Mol Cell Proteomics ; 15(2): 715-25, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26604261

ABSTRACT

AMPylation of mammalian small GTPases by bacterial virulence factors can be a key step in bacterial infection of host cells, and constitutes a potential drug target. This posttranslational modification also exists in eukaryotes, and AMP transferase activity was recently assigned to HYPE Filamentation induced by cyclic AMP domain containing protein (FICD) protein, which is conserved from Caenorhabditis elegans to humans. In contrast to bacterial AMP transferases, only a small number of HYPE substrates have been identified by immunoprecipitation and mass spectrometry approaches, and the full range of targets is yet to be determined in mammalian cells. We describe here the first example of global chemoproteomic screening and substrate validation for HYPE-mediated AMPylation in mammalian cell lysate. Through quantitative mass-spectrometry-based proteomics coupled with novel chemoproteomic tools providing MS/MS evidence of AMP modification, we identified a total of 25 AMPylated proteins, including the previously validated substrate endoplasmic reticulum (ER) chaperone BiP (HSPA5), and also novel substrates involved in pathways of gene expression, ATP biosynthesis, and maintenance of the cytoskeleton. This dataset represents the largest library of AMPylated human proteins reported to date and a foundation for substrate-specific investigations that can ultimately decipher the complex biological networks involved in eukaryotic AMPylation.


Subject(s)
Carrier Proteins/metabolism , Cyclic AMP/metabolism , Membrane Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Proteomics , Amino Acid Sequence/drug effects , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Carrier Proteins/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Host-Pathogen Interactions , Humans , Infections/genetics , Infections/microbiology , Membrane Proteins/genetics , Monomeric GTP-Binding Proteins/genetics , Nucleotidyltransferases , Protein Processing, Post-Translational/genetics , Tandem Mass Spectrometry , Transferases/metabolism
8.
Mol Cell ; 34(2): 223-33, 2009 Apr 24.
Article in English | MEDLINE | ID: mdl-19394299

ABSTRACT

Rho family GTPases are important cellular switches and control a number of physiological functions. Understanding the molecular basis of interaction of these GTPases with their effectors is crucial in understanding their functions in the cell. Here we present the crystal structure of the complex of Rac2 bound to the split pleckstrin homology (spPH) domain of phospholipase C-gamma(2) (PLCgamma(2)). Based on this structure, we illustrate distinct requirements for PLCgamma(2) activation by Rac and EGF and generate Rac effector mutants that specifically block activation of PLCgamma(2), but not the related PLCbeta(2) isoform. Furthermore, in addition to the complex, we report the crystal structures of free spPH and Rac2 bound to GDP and GTPgammaS. These structures illustrate a mechanism of conformational switches that accompany formation of signaling active complexes and highlight the role of effector binding as a common feature of Rac and Cdc42 interactions with a variety of effectors.


Subject(s)
Phospholipase C gamma/chemistry , rac GTP-Binding Proteins/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Enzyme Activation , Epidermal Growth Factor/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Phospholipase C gamma/metabolism , Protein Interaction Mapping , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity , Thermodynamics , rac GTP-Binding Proteins/metabolism , RAC2 GTP-Binding Protein
9.
Trends Biochem Sci ; 36(2): 88-96, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20870410

ABSTRACT

Phosphoinositide-specific phospholipase C (PLC) enzymes are common signalling components linked to the activation of most cellular receptors. All PLC families are complex, modular, multi-domain proteins and together cover a broad spectrum of regulatory interactions, including direct binding to G protein subunits, small GTPases from Rho and Ras families, receptor and non-receptor tyrosine kinases and lipid components of cellular membranes. Recent structural determinations of PLC components and their complexes with regulatory proteins and direct mechanistic studies, together with earlier work, have provided the foundation to propose molecular mechanisms that stringently regulate PLC activity.


Subject(s)
Phosphoinositide Phospholipase C/metabolism , Animals , Cell Membrane/enzymology , Enzyme Activation , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Humans , Models, Molecular , Phosphoinositide Phospholipase C/chemistry , Signal Transduction
10.
Am J Hum Genet ; 91(4): 713-20, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-23000145

ABSTRACT

Whole-exome sequencing was performed in a family affected by dominantly inherited inflammatory disease characterized by recurrent blistering skin lesions, bronchiolitis, arthralgia, ocular inflammation, enterocolitis, absence of autoantibodies, and mild immunodeficiency. Exome data from three samples, including the affected father and daughter and unaffected mother, were filtered for the exclusion of reported variants, along with benign variants, as determined by PolyPhen-2. A total of eight transcripts were identified as possible candidate genes. We confirmed a variant, c.2120C>A (p.Ser707Tyr), within PLCG2 as the only de novo variant that was present in two affected family members and not present in four unaffected members. PLCG2 encodes phospholipase Cγ2 (PLCγ2), an enzyme with a critical regulatory role in various immune and inflammatory pathways. The p.Ser707Tyr substitution is located in an autoinhibitory SH2 domain that is crucial for PLCγ2 activation. Overexpression of the altered p.Ser707Tyr protein and ex vivo experiments using affected individuals' leukocytes showed clearly enhanced PLCγ2 activity, suggesting increased intracellular signaling in the PLCγ2-mediated pathway. Recently, our laboratory identified in individuals with cold-induced urticaria and immune dysregulation PLCG2 exon-skipping mutations resulting in protein products with constitutive phospholipase activity but with reduced intracellular signaling at physiological temperatures. In contrast, the p.Ser707Tyr substitution in PLCγ2 causes a distinct inflammatory phenotype that is not provoked by cold temperatures and that has different end-organ involvement and increased intracellular signaling at physiological temperatures. Our results highlight the utility of exome-sequencing technology in finding causal mutations in nuclear families with dominantly inherited traits otherwise intractable by linkage analysis.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Immunologic Deficiency Syndromes/genetics , Mutation, Missense , Phospholipase C gamma/genetics , Exome/genetics , Exons , Female , Genetic Linkage , Genetic Predisposition to Disease , Hereditary Autoinflammatory Diseases/enzymology , Hereditary Autoinflammatory Diseases/metabolism , Humans , Immunologic Deficiency Syndromes/enzymology , Immunologic Deficiency Syndromes/metabolism , Leukocytes/metabolism , Male , src Homology Domains/genetics
11.
N Engl J Med ; 366(4): 330-8, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22236196

ABSTRACT

BACKGROUND: Mendelian analysis of disorders of immune regulation can provide insight into molecular pathways associated with host defense and immune tolerance. METHODS: We identified three families with a dominantly inherited complex of cold-induced urticaria, antibody deficiency, and susceptibility to infection and autoimmunity. Immunophenotyping methods included flow cytometry, analysis of serum immunoglobulins and autoantibodies, lymphocyte stimulation, and enzymatic assays. Genetic studies included linkage analysis, targeted Sanger sequencing, and next-generation whole-genome sequencing. RESULTS: Cold urticaria occurred in all affected subjects. Other, variable manifestations included atopy, granulomatous rash, autoimmune thyroiditis, the presence of antinuclear antibodies, sinopulmonary infections, and common variable immunodeficiency. Levels of serum IgM and IgA and circulating natural killer cells and class-switched memory B cells were reduced. Linkage analysis showed a 7-Mb candidate interval on chromosome 16q in one family, overlapping by 3.5 Mb a disease-associated haplotype in a smaller family. This interval includes PLCG2, encoding phospholipase Cγ(2) (PLCγ(2)), a signaling molecule expressed in B cells, natural killer cells, and mast cells. Sequencing of complementary DNA revealed heterozygous transcripts lacking exon 19 in two families and lacking exons 20 through 22 in a third family. Genomic sequencing identified three distinct in-frame deletions that cosegregated with disease. These deletions, located within a region encoding an autoinhibitory domain, result in protein products with constitutive phospholipase activity. PLCG2-expressing cells had diminished cellular signaling at 37°C but enhanced signaling at subphysiologic temperatures. CONCLUSIONS: Genomic deletions in PLCG2 cause gain of PLCγ(2) function, leading to signaling abnormalities in multiple leukocyte subsets and a phenotype encompassing both excessive and deficient immune function. (Funded by the National Institutes of Health Intramural Research Programs and others.).


Subject(s)
Autoimmune Diseases/genetics , Cryopyrin-Associated Periodic Syndromes/genetics , Immunologic Deficiency Syndromes/genetics , Phospholipase C gamma/genetics , Sequence Deletion , Cold Temperature/adverse effects , DNA, Complementary/analysis , DNA, Complementary/isolation & purification , Female , Humans , Male , Pedigree , Phenotype , Phospholipase C gamma/metabolism , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
12.
Nat Genet ; 38(12): 1397-405, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17086182

ABSTRACT

Nephrotic syndrome, a malfunction of the kidney glomerular filter, leads to proteinuria, edema and, in steroid-resistant nephrotic syndrome, end-stage kidney disease. Using positional cloning, we identified mutations in the phospholipase C epsilon gene (PLCE1) as causing early-onset nephrotic syndrome with end-stage kidney disease. Kidney histology of affected individuals showed diffuse mesangial sclerosis (DMS). Using immunofluorescence, we found PLCepsilon1 expression in developing and mature glomerular podocytes and showed that DMS represents an arrest of normal glomerular development. We identified IQ motif-containing GTPase-activating protein 1 as a new interaction partner of PLCepsilon1. Two siblings with a missense mutation in an exon encoding the PLCepsilon1 catalytic domain showed histology characteristic of focal segmental glomerulosclerosis. Notably, two other affected individuals responded to therapy, making this the first report of a molecular cause of nephrotic syndrome that may resolve after therapy. These findings, together with the zebrafish model of human nephrotic syndrome generated by plce1 knockdown, open new inroads into pathophysiology and treatment mechanisms of nephrotic syndrome.


Subject(s)
Mutation , Nephrotic Syndrome/enzymology , Nephrotic Syndrome/genetics , Type C Phospholipases/genetics , Animals , Child , Child, Preschool , Cloning, Molecular , Disease Models, Animal , Female , Gene Targeting , Genes, Recessive , Homozygote , Humans , Infant , Kidney/enzymology , Kidney/pathology , Male , Models, Genetic , Mutation, Missense , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/pathology , Phosphoinositide Phospholipase C , Rats , Sequence Deletion , Zebrafish/genetics
13.
Biochim Biophys Acta Mol Basis Dis ; 1869(2): 166601, 2023 02.
Article in English | MEDLINE | ID: mdl-36442790

ABSTRACT

BACKGROUND: Development of adult T-cell leukemia/lymphoma (ATL) involves human T-cell leukemia virus type 1 (HTLV-1) infection and accumulation of somatic mutations. The most frequently mutated gene in ATL (36 % of cases) is phospholipase C gamma1 (PLCG1). PLCG1 is also frequently mutated in other T-cell lymphomas. However, the functional consequences of the PLCG1 mutations in cancer cells have not been characterized. METHODS: We compared the activity of the wild-type PLCγ1 with that of a mutant carrying a hot-spot mutation of PLCγ1 (S345F) observed in ATL, both in cells and in cell-free assays. To analyse the impact of the mutation on cellular properties, we quantified cellular proliferation, aggregation, chemotaxis and apoptosis by live cell-imaging in an S345F+ ATL-derived cell line (KK1) and a KK1 cell line in which we reverted the mutation to the wild-type sequence using CRISPR/Cas9 and homology-directed repair. FINDINGS: The PLCγ1 S345F mutation results in an increase of basal PLC activity in vitro and in different cell types. This higher basal activity is further enhanced by upstream signalling. Reversion of the S345F mutation in the KK1 cell line resulted in reduction of the PLC activity, lower rates of proliferation and aggregation, and a marked reduction in chemotaxis towards CCL22. The PLCγ1-pathway inhibitors ibrutinib and ritonavir reduced both the PLC activity and the tested functions of KK1 cells. INTERPRETATION: Consistent with observations from clinical studies, our data provide direct evidence that activated variants of the PLCγ1 enzyme contribute to the properties of the malignant T-cell clone in ATL. FUNDING: MRC (UK) Project Grant (P028160).


Subject(s)
Human T-lymphotropic virus 1 , Leukemia-Lymphoma, Adult T-Cell , Phospholipase C gamma , Adult , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Mutation , Phospholipase C gamma/genetics
14.
Sci Adv ; 8(25): eabp9688, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35749497

ABSTRACT

PLCγ enzymes are autoinhibited in resting cells and form key components of intracellular signaling that are also linked to disease development. Insights into physiological and aberrant activation of PLCγ require understanding of an active, membrane-bound form, which can hydrolyze inositol-lipid substrates. Here, we demonstrate that PLCγ1 cannot bind membranes unless the autoinhibition is disrupted. Through extensive molecular dynamics simulations and experimental evidence, we characterize membrane binding by the catalytic core domains and reveal previously unknown sites of lipid interaction. The identified sites act in synergy, overlap with autoinhibitory interfaces, and are shown to be critical for the phospholipase activity in cells. This work provides direct evidence that PLCγ1 is inhibited through obstruction of its membrane-binding surfaces by the regulatory region and that activation must shift PLCγ1 to a conformation competent for membrane binding. Knowledge of the critical sites of membrane interaction extends the mechanistic framework for activation, dysregulation, and therapeutic intervention.


Subject(s)
Lipids , Signal Transduction , Catalytic Domain
15.
Mol Neurodegener ; 16(1): 22, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33823896

ABSTRACT

The central role of the resident innate immune cells of the brain (microglia) in neurodegeneration has become clear over the past few years largely through genome-wide association studies (GWAS), and has rapidly become an active area of research. However, a mechanistic understanding (gene to function) has lagged behind. That is now beginning to change, as exemplified by a number of recent exciting and important reports that provide insight into the function of two key gene products - TREM2 (Triggering Receptor Expressed On Myeloid Cells 2) and PLCγ2 (Phospholipase C gamma2) - in microglia, and their role in neurodegenerative disorders. In this review we explore and discuss these recent advances and the opportunities that they may provide for the development of new therapies.


Subject(s)
Alzheimer Disease/immunology , Connective Tissue Cells/metabolism , Lymphocytes/metabolism , Membrane Glycoproteins/physiology , Microglia/metabolism , Myeloid Cells/metabolism , Phospholipase C gamma/physiology , Receptors, Immunologic/physiology , Signal Transduction/physiology , Age of Onset , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Humans , Lipid Metabolism , Membrane Glycoproteins/chemistry , Microglia/physiology , Models, Molecular , Mutation , Phospholipase C gamma/chemistry , Phospholipase C gamma/genetics , Protein Conformation , Protein Domains , Protein Interaction Mapping , Receptors, Immunologic/chemistry , Sequence Homology, Amino Acid , Structure-Activity Relationship
16.
J Biol Chem ; 284(34): 23083-93, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19531496

ABSTRACT

Phospholipase C gamma isozymes (PLC gamma 1 and PLC gamma 2) have a crucial role in the regulation of a variety of cellular functions. Both enzymes have also been implicated in signaling events underlying aberrant cellular responses. Using N-ethyl-N-nitrosourea (ENU) mutagenesis, we have recently identified single point mutations in murine PLC gamma 2 that lead to spontaneous inflammation and autoimmunity. Here we describe further, mechanistic characterization of two gain-of-function mutations, D993G and Y495C, designated as ALI5 and ALI14. The residue Asp-993, mutated in ALI5, is a conserved residue in the catalytic domain of PLC enzymes. Analysis of PLC gamma 1 and PLC gamma 2 with point mutations of this residue showed that removal of the negative charge enhanced PLC activity in response to EGF stimulation or activation by Rac. Measurements of PLC activity in vitro and analysis of membrane binding have suggested that ALI5-type mutations facilitate membrane interactions without compromising substrate binding and hydrolysis. The residue mutated in ALI14 (Tyr-495) is within the spPH domain. Replacement of this residue had no effect on folding of the domain and enhanced Rac activation of PLC gamma 2 without increasing Rac binding. Importantly, the activation of the ALI14-PLC gamma 2 and corresponding PLC gamma 1 variants was enhanced in response to EGF stimulation and bypassed the requirement for phosphorylation of critical tyrosine residues. ALI5- and ALI14-type mutations affected basal activity only slightly; however, their combination resulted in a constitutively active PLC. Based on these data, we suggest that each mutation could compromise auto-inhibition in the inactive PLC, facilitating the activation process; in addition, ALI5-type mutations could enhance membrane interaction in the activated state.


Subject(s)
Mutation/genetics , Phospholipase C gamma/chemistry , Phospholipase C gamma/metabolism , Alkylating Agents/pharmacology , Amino Acid Sequence , Animals , COS Cells , Calorimetry , Catalytic Domain , Cell Line , Chlorocebus aethiops , Drosophila , Ethylnitrosourea/pharmacology , Isoenzymes , Magnetic Resonance Spectroscopy , Mice , Molecular Sequence Data , Mutation/drug effects , Phospholipase C gamma/genetics , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Sequence Homology, Amino Acid , Structure-Activity Relationship , Surface Plasmon Resonance
17.
EBioMedicine ; 51: 102607, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31918402

ABSTRACT

BACKGROUND: PLCγ enzymes are key nodes in cellular signal transduction and their mutated and rare variants have been recently implicated in development of a range of diseases with unmet need including cancer, complex immune disorders, inflammation and neurodegenerative diseases. However, molecular nature of activation and the impact and dysregulation mechanisms by mutations, remain unclear; both are critically dependent on comprehensive characterization of the intact PLCγ enzymes. METHODS: For structural studies we applied cryo-EM, cross-linking mass spectrometry and hydrogen-deuterium exchange mass spectrometry. In parallel, we compiled mutations linked to main pathologies, established their distribution and assessed their impact in cells and in vitro. FINDINGS: We define structure of a complex containing an intact, autoinhibited PLCγ1 and the intracellular part of FGFR1 and show that the interaction is centred on the nSH2 domain of PLCγ1. We define the architecture of PLCγ1 where an autoinhibitory interface involves the cSH2, spPH, TIM-barrel and C2 domains; this relative orientation occludes PLCγ1 access to its substrate. Based on this framework and functional characterization, the mechanism leading to an increase in PLCγ1 activity for the largest group of mutations is consistent with the major, direct impact on the autoinhibitory interface. INTERPRETATION: We reveal features of PLCγ enzymes that are important for determining their activation status. Targeting such features, as an alternative to targeting the PLC active site that has so far not been achieved for any PLC, could provide new routes for clinical interventions related to various pathologies driven by PLCγ deregulation. FUND: CR UK, MRC and AstaZeneca.


Subject(s)
Mutation/genetics , Phospholipase C gamma/chemistry , Phospholipase C gamma/genetics , Humans , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Phospholipase C gamma/ultrastructure , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/metabolism
18.
Methods Mol Biol ; 462: 379-89, 2009.
Article in English | MEDLINE | ID: mdl-19160682

ABSTRACT

Phosphatidylinositol-specific phospholipase C (PLC) enzymes catalyze the hydrolysis of phophatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] to diacylglycerol (DAG) and inositol 1,4,5-triphosphate [Ins(1,4,5)P3]. PLCepsilon is a recently discovered isoform that has been shown to be activated by members of the Ras and Rho families of guanosine trisphosphatases (GTPases) as well as subunits of heterotrimeric G-proteins. We describe a method for expressing a truncated PLCepsilon variant as an MBP fusion protein in E. coli. Subsequently, we describe the methodology necessary to reconstitute this protein with K-Ras-4B and RhoA GTPases and measure its activation.


Subject(s)
Baculoviridae/enzymology , Phosphoinositide Phospholipase C/isolation & purification , Phosphoinositide Phospholipase C/metabolism , ras Proteins/isolation & purification , ras Proteins/pharmacology , rho GTP-Binding Proteins/isolation & purification , rho GTP-Binding Proteins/pharmacology , Animals , Cell Line , Cell Membrane/chemistry , Cell-Free System , Chromatography, Affinity , Chromatography, Gel , Enzyme Activation/drug effects , Escherichia coli/cytology , Isoenzymes/genetics , Isoenzymes/isolation & purification , Isoenzymes/metabolism , Phosphoinositide Phospholipase C/genetics , Rats , Sequence Deletion , Solubility
19.
Biophys J ; 95(10): L69-71, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18757561

ABSTRACT

We report what to our knowledge is a novel approach for simultaneous imaging of two different Förster resonance energy transfer (FRET) sensors in the same cell with minimal spectral cross talk. Previous methods based on spectral ratiometric imaging of the two FRET sensors have been limited by the availability of suitably bright acceptors for the second FRET pair and the spectral cross talk incurred when measuring in four spectral windows. In contrast to spectral ratiometric imaging, fluorescence lifetime imaging (FLIM) requires measurement of the donor fluorescence only and is independent of emission from the acceptor. By combining FLIM-FRET of the novel red-shifted TagRFP/mPlum FRET pair with spectral ratiometric imaging of an ECFP/Venus pair we were thus able to maximize the spectral separation between our chosen fluorophores while at the same time overcoming the low quantum yield of the far red acceptor mPlum. Using this technique, we could read out a TagRFP/mPlum intermolecular FRET sensor for reporting on small Ras GTP-ase activation in live cells after epidermal growth factor stimulation and an ECFP/Venus Cameleon FRET sensor for monitoring calcium transients within the same cells. The combination of spectral ratiometric imaging of ECFP/Venus and high-speed FLIM-FRET of TagRFP/mPlum can thus increase the spectral bandwidth available and provide robust imaging of multiple FRET sensors within the same cell. Furthermore, since FLIM does not require equal stoichiometries of donor and acceptor, this approach can be used to report on both unimolecular FRET biosensors and protein-protein interactions with the same cell.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Fluorescence Resonance Energy Transfer/instrumentation , Protein Interaction Mapping/instrumentation , Signal Transduction/physiology , Animals , COS Cells , Chlorocebus aethiops , Equipment Design , Equipment Failure Analysis , Fluorescence Resonance Energy Transfer/methods , Protein Interaction Mapping/methods
20.
Biomol NMR Assign ; 12(2): 231-235, 2018 10.
Article in English | MEDLINE | ID: mdl-29582384

ABSTRACT

Fibroblast growth factors receptors (FGFR) are transmembrane protein tyrosine kinases involved in many cellular process, including growth, differentiation and angiogenesis. Dysregulation of FGFR enzymatic activity is associated with developmental disorders and cancers; therefore FGFRs have become attractive targets for drug discovery, with a number of agents in late-stage clinical trials. Here, we present the backbone resonance assignments of FGFR3 tyrosine kinase domain in the ligand-free form and in complex with the canonical FGFR kinase inhibitor PD173074. Analysis of chemical shift changes upon inhibitor binding highlights a characteristic pattern of allosteric network perturbations that is of relevance for future drug discovery activities aimed at development of conformationally-selective FGFR inhibitors.


Subject(s)
Apoproteins/chemistry , Apoproteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Pyrimidines/metabolism , Receptor, Fibroblast Growth Factor, Type 3/chemistry , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Apoproteins/antagonists & inhibitors , Humans , Protein Binding , Protein Domains , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL