89 results on '"Casarotto MG"'
Search Results
2. Effects of an alpha-helical ryanodine receptor C-terminal tail peptide on ryanodine receptor activity: Modulation by Homer
- Author
-
Pouliquin, P, Pace, Sm, Curtis, Sm, Harvey, Pj, Gallant, Em, Zorzato, Francesco, Casarotto, Mg, and Dulhunty, Af
- Subjects
ryanodine receptor ,calcium regulation - Published
- 2006
3. Structural model of human IL-13 defines the spatial interactions with the IL-13Rα/IL-4Rα receptor.
- Author
-
Casarotto, MG, Zuegg, J, Webb, DC, and Foster, PS
- Subjects
- *
INTERLEUKINS , *CYTOKINESIS - Abstract
Summary Interleukin-13 (IL-13) plays a key role in immune responses and inflammation. A structural model of human IL-13 (HuIL-13) based on the nuclear magnetic resonance and X-ray structure of IL-4 is put forward. Unlike previous models, this model is based on new sequence alignments that take into account the formation of the two disulfide linkages that have been determined experimentally. The proposed structure of human IL-13 is similar to IL-4, consisting of a four helix bundle with hydrophobic residues lining the core of the molecule and surface polar residues showing a high degree of solvent accessibility. Regions of HuIL-13 that are critical for the interaction with its receptors are explored and discussed in relation to existing mutagenic studies. From these studies we predict that helices A and C of HuIL-13 interact with the IL-4 receptor alpha (IL-4Rα) region and helix D is responsible for the interaction with the IL-13 receptor alpha 1 (IL-13Rα1) receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2001
- Full Text
- View/download PDF
4. Complex Actions of FKBP12 on RyR1 Ion Channel Activity Consistent with Negative Co-Operativity in FKBP12 Binding to the RyR1 Tetramer.
- Author
-
Richardson SJ, Thekkedam CG, Casarotto MG, Beard NA, and Dulhunty AF
- Subjects
- Animals, Rabbits, Muscle, Skeletal metabolism, Calcium metabolism, Ryanodine Receptor Calcium Release Channel metabolism, Tacrolimus Binding Protein 1A metabolism, Protein Binding, Sarcoplasmic Reticulum metabolism
- Abstract
The association of the 12 KDa FK506 binding protein (FKBP12) with ryanodine receptor type 1 (RyR1) in skeletal muscle is thought to suppress RyR1 channel opening and contribute to healthy muscle function. The strongest evidence for this role is increased RyR1 channel activity following FKBP12 dissociation. However, the corollary that channel activity will decrease when FKBP12 is added back to FKBP12-depleted RyR1 is not well established, and when reported, the time- and concentration-dependence of inhibition vary over orders of magnitude. Here, we address this problem with an investigation of the molecular mechanisms of the FKBP12 regulation of RyR1. Muscle processing to obtain sarcoplasmic reticulum (SR) vesicle preparations enriched in RyR1 resulted in substantial FKBP12 dissociation from RyR1, indicating low-affinity binding. Conversely, high-affinity binding was indicated by some FKBP12 remaining bound to RyR1 after solubilization. We report, for the first time, an increase in the activity of FKBP12-depleted channels after the addition of exogenous FKBP12 (5 nM to 5 µM), followed by a reduction in activity consistent with inhibition after 20-30 min exposure to higher [FKBP12]s. Both the increase and later decline in activity were time- and concentration-dependent. The results suggest a high-affinity activation when FKBP12 binding sites on the RyR1 tetramer are partially occupied by FKBP12 and lower affinity inhibition as more RyR1 monomers become occupied. These novel results imply negative cooperativity in FKBP12 binding to RyR1 and a dynamic role for FKBP12/RyR1 interactions in intact muscle fibers.
- Published
- 2025
- Full Text
- View/download PDF
5. Transcriptional reprogramming by mutated IRF4 in lymphoma.
- Author
-
Schleussner N, Cauchy P, Franke V, Giefing M, Fornes O, Vankadari N, Assi SA, Costanza M, Weniger MA, Akalin A, Anagnostopoulos I, Bukur T, Casarotto MG, Damm F, Daumke O, Edginton-White B, Gebhardt JCM, Grau M, Grunwald S, Hansmann ML, Hartmann S, Huber L, Kärgel E, Lusatis S, Noerenberg D, Obier N, Pannicke U, Fischer A, Reisser A, Rosenwald A, Schwarz K, Sundararaj S, Weilemann A, Winkler W, Xu W, Lenz G, Rajewsky K, Wasserman WW, Cockerill PN, Scheidereit C, Siebert R, Küppers R, Grosschedl R, Janz M, Bonifer C, and Mathas S
- Subjects
- Humans, B-Lymphocytes metabolism, DNA, Gene Expression Regulation, Interferon Regulatory Factors genetics, Interferon Regulatory Factors metabolism, Lymphoma genetics
- 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., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
6. FKBP12 binds to the cardiac ryanodine receptor with negative cooperativity: implications for heart muscle physiology in health and disease.
- Author
-
Richardson SJ, Thekkedam CG, Casarotto MG, Beard NA, and Dulhunty AF
- Subjects
- Humans, Animals, Sheep, Myocardium metabolism, Calcium Signaling, Protein Isoforms metabolism, Protein Isoforms pharmacology, Calcium metabolism, Tacrolimus Binding Protein 1A metabolism, Tacrolimus Binding Protein 1A pharmacology, Ryanodine Receptor Calcium Release Channel chemistry, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Cardiac ryanodine receptors (RyR2) release the Ca
2+ from intracellular stores that is essential for cardiac myocyte contraction. The ion channel opening is tightly regulated by intracellular factors, including the FK506 binding proteins, FKBP12 and FKBP12.6. The impact of these proteins on RyR2 activity and cardiac contraction is debated, with often apparently contradictory experimental results, particularly for FKBP12. The isoform that regulates RyR2 has generally been considered to be FKBP12.6, despite the fact that FKBP12 is the major isoform associated with RyR2 in some species and is bound in similar proportions to FKBP12.6 in others, including sheep and humans. Here, we show time- and concentration-dependent effects of adding FKBP12 to RyR2 channels that were partly depleted of FKBP12/12.6 during isolation. The added FKBP12 displaced most remaining endogenous FKBP12/12.6. The results suggest that FKBP12 activates RyR2 with high affinity and inhibits RyR2 with lower affinity, consistent with a model of negative cooperativity in FKBP12 binding to each of the four subunits in the RyR tetramer. The easy dissociation of some FKBP12/12.6 could dynamically alter RyR2 activity in response to changes in in vivo regulatory factors, indicating a significant role for FKBP12/12.6 in Ca2+ signalling and cardiac function in healthy and diseased hearts. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.- Published
- 2023
- Full Text
- View/download PDF
7. A multimorphic mutation in IRF4 causes human autosomal dominant combined immunodeficiency.
- Author
-
Fornes O, Jia A, Kuehn HS, Min Q, Pannicke U, Schleussner N, Thouenon R, Yu Z, de Los Angeles Astbury M, Biggs CM, Galicchio M, Garcia-Campos JA, Gismondi S, Gonzalez Villarreal G, Hildebrand KJ, Hönig M, Hou J, Moshous D, Pittaluga S, Qian X, Rozmus J, Schulz AS, Staines-Boone AT, Sun B, Sun J, Uwe S, Venegas-Montoya E, Wang W, Wang X, Ying W, Zhai X, Zhou Q, Akalin A, André I, Barth TFE, Baumann B, Brüstle A, Burgio G, Bustamante JC, Casanova JL, Casarotto MG, Cavazzana M, Chentout L, Cockburn IA, Costanza M, Cui C, Daumke O, Del Bel KL, Eibel H, Feng X, Franke V, Gebhardt JCM, Götz A, Grunwald S, Hoareau B, Hughes TR, Jacobsen EM, Janz M, Jolma A, Lagresle-Peyrou C, Lai N, Li Y, Lin S, Lu HY, Lugo-Reyes SO, Meng X, Möller P, Moreno-Corona N, Niemela JE, Novakovsky G, Perez-Caraballo JJ, Picard C, Poggi L, Puig-Lombardi ME, Randall KL, Reisser A, Schmitt Y, Seneviratne S, Sharma M, Stoddard J, Sundararaj S, Sutton H, Tran LQ, Wang Y, Wasserman WW, Wen Z, Winkler W, Xiong E, Yang AWH, Yu M, Zhang L, Zhang H, Zhao Q, Zhen X, Enders A, Kracker S, Martinez-Barricarte R, Mathas S, Rosenzweig SD, Schwarz K, Turvey SE, and Wang JY
- Subjects
- Mice, Animals, Humans, B-Lymphocytes, DNA metabolism, Mutation, Interferon Regulatory Factors, Gene Expression Regulation
- Abstract
Interferon regulatory factor 4 (IRF4) is a transcription factor (TF) and key regulator of immune cell development and function. We report a recurrent heterozygous mutation in IRF4, p.T95R, causing an autosomal dominant combined immunodeficiency (CID) in seven patients from six unrelated families. The patients exhibited profound susceptibility to opportunistic infections, notably Pneumocystis jirovecii , and presented with agammaglobulinemia. Patients' B cells showed impaired maturation, decreased immunoglobulin isotype switching, and defective plasma cell differentiation, whereas their T cells contained reduced T
H 17 and TFH populations and exhibited decreased cytokine production. A knock-in mouse model of heterozygous T95R showed a severe defect in antibody production both at the steady state and after immunization with different types of antigens, consistent with the CID observed in these patients. The IRF4T95R variant maps to the TF's DNA binding domain, alters its canonical DNA binding specificities, and results in a simultaneous multimorphic combination of loss, gain, and new functions for IRF4. IRF4T95R behaved as a gain-of-function hypermorph by binding to DNA with higher affinity than IRF4WT . Despite this increased affinity for DNA, the transcriptional activity on IRF4 canonical genes was reduced, showcasing a hypomorphic activity of IRF4T95R . Simultaneously, IRF4T95R functions as a neomorph by binding to noncanonical DNA sites to alter the gene expression profile, including the transcription of genes exclusively induced by IRF4T95R but not by IRF4WT . This previously undescribed multimorphic IRF4 pathophysiology disrupts normal lymphocyte biology, causing human disease.- Published
- 2023
- Full Text
- View/download PDF
8. A maladaptive feedback mechanism between the extracellular matrix and cytoskeleton contributes to hypertrophic cardiomyopathy pathophysiology.
- Author
-
Viola HM, Richworth C, Solomon T, Chin IL, Szappanos HC, Sundararaj S, Shishmarev D, Casarotto MG, Choi YS, and Hool LC
- Subjects
- Mice, Humans, Animals, Feedback, Cytoskeleton metabolism, Myocytes, Cardiac metabolism, Troponin I genetics, Troponin I metabolism, Extracellular Matrix metabolism, Hydrogels, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism
- 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., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
9. Nuclear Transport of Respiratory Syncytial Virus Matrix Protein Is Regulated by Dual Phosphorylation Sites.
- Author
-
Ghildyal R, Teng MN, Tran KC, Mills J, Casarotto MG, Bardin PG, and Jans DA
- Subjects
- Active Transport, Cell Nucleus, Aged, Cell Nucleus metabolism, Cytoplasm metabolism, Humans, Phosphorylation, Respiratory Syncytial Virus, Human
- Abstract
Respiratory syncytial virus (RSV) is a major cause of respiratory infections in infants and the elderly. Although the RSV matrix (M) protein has key roles in the nucleus early in infection, and in the cytoplasm later, the molecular basis of switching between the nuclear and cytoplasmic compartments is not known. Here, we show that protein kinase CK2 can regulate M nucleocytoplasmic distribution, whereby inhibition of CK2 using the specific inhibitor 4,5,6,7-tetrabromobenzo-triazole (TBB) increases M nuclear accumulation in infected cells as well as when ectopically expressed in transfected cells. We use truncation/mutagenic analysis for the first time to show that serine (S) 95 and threonine (T) 205 are key CK2 sites that regulate M nuclear localization. Dual alanine (A)-substitution to prevent phosphorylation abolished TBB- enhancement of nuclear accumulation, while aspartic acid (D) substitution to mimic phosphorylation at S95 increased nuclear accumulation. D95 also induced cytoplasmic aggregate formation, implying that a negative charge at S95 may modulate M oligomerization. A95/205 substitution in recombinant RSV resulted in reduced virus production compared with wild type, with D95/205 substitution resulting in an even greater level of attenuation. Our data support a model where unphosphorylated M is imported into the nucleus, followed by phosphorylation of T205 and S95 later in infection to facilitate nuclear export and cytoplasmic retention of M, respectively, as well as oligomerization/virus budding. In the absence of widely available, efficacious treatments to protect against RSV, the results raise the possibility of antiviral strategies targeted at CK2.
- Published
- 2022
- Full Text
- View/download PDF
10. Molecular interactions of STAC proteins with skeletal muscle dihydropyridine receptor and excitation-contraction coupling.
- Author
-
Shishmarev D, Rowland E, Aditya S, Sundararaj S, Oakley AJ, Dulhunty AF, and Casarotto MG
- Subjects
- Excitation Contraction Coupling physiology, Muscle, Skeletal physiology, Protein Isoforms metabolism, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type genetics, Calcium Channels, L-Type metabolism, Ryanodine Receptor Calcium Release Channel
- 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 Ca
2+ 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., (© 2022 The Authors. Protein Science published by Wiley Periodicals LLC on behalf of The Protein Society.)- Published
- 2022
- Full Text
- View/download PDF
11. The molecular basis for the development of adult T-cell leukemia/lymphoma in patients with an IRF4 K59R mutation.
- Author
-
Sundararaj S, Seneviratne S, Williams SJ, Enders A, and Casarotto MG
- Subjects
- Adult, Humans, Mutation, 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
- 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 (IRF4
K59R ) 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., (© 2021 The Protein Society.)- Published
- 2022
- Full Text
- View/download PDF
12. The Antiviral Drug Efavirenz in Breast Cancer Stem Cell Therapy.
- Author
-
Chiou PT, Ohms S, Board PG, Dahlstrom JE, Rangasamy D, and Casarotto MG
- Abstract
Although many breast cancer therapies show initial success in the treatment of the primary tumour, they often fail to eliminate a sub-population of cells known as cancer stem cells (CSCs). These cells are recognised for their self-renewal properties and for their capacity for differentiation often leading to chemo/radio-resistance. The antiviral drug Efavirenz has been shown to be effective in eliminating triple-negative breast cancer cells, and here we examine its effect on breast CSCs. The effects of Efavirenz on CSCs for several breast cancer cell lines were investigated by examining cellular changes upon drug treatment, including CSC numbers, morphology, RNA/microRNA expression and levels of epithelial/mesenchymal CSC subtypes. Efavirenz treatment resulted in a decrease in the size and number of tumorspheres and a reduction in epithelial-type CSC levels, but an increase in mesenchymal-type CSCs. Efavirenz caused upregulation of several CSC-related genes as well as miR-21 , a CSC marker and miR-182 , a CSC suppressor gene. We conclude that Efavirenz alters the phenotype and expression of key genes in breast CSCs, which has important potential therapeutic implications.
- Published
- 2021
- Full Text
- View/download PDF
13. Molecular interactions of IRF4 in B cell development and malignancies.
- Author
-
Sundararaj S and Casarotto MG
- 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., Competing Interests: Conflict of interestThe authors declare no competing interests., (© International Union for Pure and Applied Biophysics (IUPAB) and Springer-Verlag GmbH Germany, part of Springer Nature 2021.)
- Published
- 2021
- Full Text
- View/download PDF
14. Glutathione transferase Omega 1 confers protection against azoxymethane-induced colorectal tumour formation.
- Author
-
Tummala P, Rooke M, Dahlstrom JE, Takahashi S, Casarotto MG, Fernando N, Hughes MM, O'Neill LAJ, and Board PG
- Subjects
- Animals, Carcinogens toxicity, Colitis chemically induced, Colorectal Neoplasms etiology, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Dextran Sulfate toxicity, Inflammation etiology, Inflammation metabolism, Inflammation pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Azoxymethane toxicity, Carrier Proteins physiology, Colitis complications, Colorectal Neoplasms prevention & control, Glutathione Transferase physiology, Inflammation prevention & control, Interleukin-18 blood, Interleukin-1beta blood
- Abstract
Inflammatory bowel disease (IBD) is characterized by multiple alterations in cytokine expression and is a risk factor for colon cancer. The Omega class glutathione transferase GSTO1-1 regulates the release of the pro-inflammatory cytokines interleukin 1β (IL-1β) and interleukin 18 (IL-18) by deglutathionylating NEK7 in the NLRP3 inflammasome. When treated with azoxymethane and dextran sodium sulphate (AOM/DSS) as a model of IBD, Gsto1-/- mice were highly sensitive to colitis and showed a significant increase in the size and number of colon tumours compared with wild-type (WT) mice. Gsto1-/- mice treated with AOM/DSS had significantly lower serum IL-1β and IL-18 levels as well as significantly decreased interferon (IFN)-γ, decreased pSTAT1 and increased pSTAT3 levels in the distal colon compared with similarly treated WT mice. Histologically, AOM/DSS treated Gsto1-/- mice showed increased active chronic inflammation with macrophage infiltration, epithelial dysplasia and invasive adenocarcinoma compared with AOM/DSS treated WT mice. Thus, this study shows that GSTO1-1 regulates IL-1β and IL-18 activation and protects against colorectal cancer formation in the AOM/DSS model of IBD. The data suggest that while GSTO1-1 is a new target for the regulation of the NLRP3 inflammasome-associated cytokines IL-1β and IL-18 by small molecule inhibitors, there is a possibility that anti-inflammatory drugs targeting these cytokines may potentiate colon cancer in some situations., (© The Author(s) 2021. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
15. AHNAK: The quiet giant in calcium homeostasis.
- Author
-
Sundararaj S, Ravindran A, and Casarotto MG
- Subjects
- Animals, Binding Sites physiology, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type metabolism, Humans, Calcium metabolism, Homeostasis physiology, Membrane Proteins chemistry, Membrane Proteins metabolism, Neoplasm Proteins chemistry, Neoplasm Proteins metabolism
- 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., (Copyright © 2021. Published by Elsevier Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
16. Structural determinants of the IRF4/DNA homodimeric complex.
- Author
-
Sundararaj S, Seneviratne S, Williams SJ, Enders A, and Casarotto MG
- Subjects
- 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, DNA chemistry, Interferon Regulatory Factors chemistry
- 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., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
17. Efavirenz as a potential drug for the treatment of triple-negative breast cancers.
- Author
-
Chiou PT, Ohms S, Board PG, Dahlstrom JE, Rangasamy D, and Casarotto MG
- Subjects
- Cell Death, Cell Line, Tumor, Cell Proliferation drug effects, Cell Shape drug effects, Down-Regulation, Fatty Acids metabolism, Female, Humans, Phenotype, Transcriptome, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Alkynes therapeutic use, Antineoplastic Agents therapeutic use, Benzoxazines therapeutic use, Cyclopropanes therapeutic use, Long Interspersed Nucleotide Elements, Reverse Transcriptase Inhibitors therapeutic use, Triple Negative Breast Neoplasms drug therapy
- Abstract
Purpose: In contrast to hormone receptor driven breast cancer, patients presenting with triple-negative breast cancer (TNBC) often have limited drug treatment options. Efavirenz, a non-nucleoside reverse transcriptase (RT) inhibitor targets abnormally overexpressed long interspersed nuclear element 1 (LINE-1) RT and has been shown to be a promising anticancer agent for treating prostate and pancreatic cancers. However, its effectiveness in treating patients with TNBC has not been comprehensively examined., Methods: In this study, the effect of Efavirenz on several TNBC cell lines was investigated by examining several cellular characteristics including viability, cell division and death, changes in cell morphology as well as the expression of LINE-1., Results: The results show that in a range of TNBC cell lines, Efavirenz causes cell death, retards cell proliferation and changes cell morphology to an epithelial-like phenotype. In addition, it is the first time that a whole-genome RNA sequence analysis has identified the fatty acid metabolism pathway as a key regulator in this Efavirenz-induced anticancer process., Conclusion: In summary, we propose Efavirenz is a potential anti-TNBC drug and that its mode of action can be linked to the fatty acid metabolism pathway.
- Published
- 2021
- Full Text
- View/download PDF
18. Peptide mimetic compounds can activate or inhibit cardiac and skeletal ryanodine receptors.
- Author
-
Robinson K, Culley D, Waring S, Lamb GD, Easton C, Casarotto MG, and Dulhunty AF
- Subjects
- Animals, Biomimetics, Calcium metabolism, Muscle Contraction drug effects, Muscle, Skeletal ultrastructure, Myocardium ultrastructure, Rabbits, Ryanodine Receptor Calcium Release Channel physiology, Sarcoplasmic Reticulum drug effects, Sarcoplasmic Reticulum metabolism, Scorpion Venoms, Sheep, Peptides pharmacology, Ryanodine Receptor Calcium Release Channel drug effects, Sarcoplasmic Reticulum chemistry
- Abstract
Aims: Our aim was to characterise the actions of novel BIT compounds with structures based on peptides and toxins that bind to significant regulatory sites on ryanodine receptor (RyR) Ca
2+ release channels. RyRs, located in sarcoplasmic reticulum (SR) Ca2+ store membranes of striated muscle, are essential for muscle contraction. Although severe sometimes-deadly myopathies occur when the channels become hyperactive following genetic or acquired changes, specific inhibitors of RyRs are rare., Main Methods: The effect of BIT compounds was determined by spectrophotometric analysis of Ca2+ release from isolated SR vesicles, analysis of single RyR channel activity in artificial lipid bilayers and contraction of intact and skinned skeletal muscle fibres., Key Findings: The inhibitory compounds reduced: (a) Ca2+ release from SR vesicles with IC50s of 1.1-2.5 μM, competing with activation by parent peptides and toxins; (b) single RyR ion channel activity with IC50s of 0.5-1.5 μM; (c) skinned fibre contraction. In contrast, activating BIT compounds increased Ca2+ release with an IC50 of 5.0 μM and channel activity with AC50s of 2 to 12 nM and enhanced skinned fibre contraction. Sub-conductance activity dominated channel activity with both inhibitors and activators. Effects of all compounds on skeletal and cardiac RyRs were similar and reversible. Competition experiments suggest that the BIT compounds bind to the regulatory helical domains of the RyRs that impact on channel gating mechanisms through long-range allosteric interactions., Significance: The BIT compounds are strong modulators of RyR activity and provide structural templates for novel research tools and drugs to combat muscle disease., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
19. Development of Benzenesulfonamide Derivatives as Potent Glutathione Transferase Omega-1 Inhibitors.
- Author
-
Xie Y, Tummala P, Oakley AJ, Deora GS, Nakano Y, Rooke M, Cuellar ME, Strasser JM, Dahlin JL, Walters MA, Casarotto MG, Board PG, and Baell JB
- Subjects
- Animals, Drug Development, Glutathione Transferase chemistry, Glutathione Transferase metabolism, Humans, Male, Mice, Molecular Docking Simulation, Structure-Activity Relationship, Benzenesulfonamides, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Glutathione Transferase antagonists & inhibitors, Sulfonamides chemistry, Sulfonamides pharmacology
- Abstract
Glutathione transferase omega-1 (GSTO1-1) is an enzyme whose function supports the activation of interleukin (IL)-1β and IL-18 that are implicated in a variety of inflammatory disease states for which small-molecule inhibitors are sought. The potent reactivity of the active-site cysteine has resulted in reported inhibitors that act by covalent labeling. In this study, structure-activity relationship (SAR) elaboration of the reported GSTO1-1 inhibitor C1-27 was undertaken. Compounds were evaluated for inhibitory activity toward purified recombinant GSTO1-1 and for indicators of target engagement in cell-based assays. As covalent inhibitors, the k
inact / KI values of selected compounds were determined, as well as in vivo pharmacokinetics analysis. Cocrystal structures of key novel compounds in complex with GSTO1-1 were also solved. This study represents the first application of a biochemical assay for GSTO1-1 to determine kinact / KI values for tested inhibitors and the most extensive set of cell-based data for a GSTO1-1 inhibitor SAR series reported to date. Our research culminated in the discovery of 25 , which we propose as the preferred biochemical tool to interrogate cellular responses to GSTO1-1 inhibition.- Published
- 2020
- Full Text
- View/download PDF
20. Glutathione Transferase Omega-1 Regulates NLRP3 Inflammasome Activation through NEK7 Deglutathionylation.
- Author
-
Hughes MM, Hooftman A, Angiari S, Tummala P, Zaslona Z, Runtsch MC, McGettrick AF, Sutton CE, Diskin C, Rooke M, Takahashi S, Sundararaj S, Casarotto MG, Dahlstrom JE, Palsson-McDermott EM, Corr SC, Mills KHG, Preston RJS, Neamati N, Xie Y, Baell JB, Board PG, and O'Neill LAJ
- Subjects
- Animals, Cytokines metabolism, HEK293 Cells, Humans, Inflammation metabolism, Inflammation Mediators metabolism, Mice, Mice, Inbred C57BL, Glutathione Transferase metabolism, Inflammasomes metabolism, NIMA-Related Kinases metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism
- 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)-1β 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., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
21. Recent advances in understanding the ryanodine receptor calcium release channels and their role in calcium signalling.
- Author
-
Dulhunty AF, Beard NA, and Casarotto MG
- Subjects
- Animals, Binding Sites genetics, Cryoelectron Microscopy, Humans, Protein Conformation, Ryanodine Receptor Calcium Release Channel chemistry, Ryanodine Receptor Calcium Release Channel genetics, Calcium Signaling genetics, Ryanodine Receptor Calcium Release Channel physiology
- Abstract
The ryanodine receptor calcium release channel is central to cytoplasmic Ca
2+ signalling in skeletal muscle, the heart, and many other tissues, including the central nervous system, lymphocytes, stomach, kidney, adrenal glands, ovaries, testes, thymus, and lungs. The ion channel protein is massive (more than 2.2 MDa) and has a structure that has defied detailed determination until recent developments in cryo-electron microscopy revealed much of its structure at near-atomic resolution. The availability of this high-resolution structure has provided the most significant advances in understanding the function of the ion channel in the past 30 years. We can now visualise the molecular environment of individual amino acid residues that form binding sites for essential modulators of ion channel function and determine its role in Ca2+ signalling. Importantly, the structure has revealed the structural environment of the many deletions and point mutations that disrupt Ca2+ signalling in skeletal and cardiac myopathies and neuropathies. The implications are of vital importance to our understanding of the molecular basis of the ion channel's function and for the design of therapies to counteract the effects of ryanodine receptor-associated disorders., Competing Interests: No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed.- Published
- 2018
- Full Text
- View/download PDF
22. 1 H, 13 C and 15 N backbone NMR chemical shift assignments of the C-terminal P4 domain of Ahnak.
- Author
-
Sundararaj S, Shishmarev D, Lin Y, Aditya S, and Casarotto MG
- Subjects
- Binding Sites, Calcium Channels, L-Type metabolism, Membrane Proteins metabolism, Protein Domains, Protein Subunits chemistry, Protein Subunits metabolism, Membrane Proteins chemistry, Nuclear Magnetic Resonance, Biomolecular
- 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 Ca
2+ signaling in cardiomyocytes by interacting with the β subunit of the L-type Ca2+ channel (CaV 1.2). 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 complete1 H,13 C and15 N 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 CaV 1.2-Ahnak complex.- Published
- 2018
- Full Text
- View/download PDF
23. Exploiting Peptidomimetics to Synthesize Compounds That Activate Ryanodine Receptor Calcium Release Channels.
- Author
-
Robinson K, Easton CJ, Dulhunty AF, and Casarotto MG
- Subjects
- Animals, Dose-Response Relationship, Drug, Molecular Structure, Muscle Fibers, Skeletal drug effects, Muscle Fibers, Skeletal metabolism, Peptides chemical synthesis, Peptides chemistry, Peptidomimetics chemical synthesis, Peptidomimetics chemistry, Rabbits, Sheep, Structure-Activity Relationship, Peptides pharmacology, Peptidomimetics pharmacology, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Ryanodine receptor (RyR) Ca
2+ -release channels are essential for contraction in skeletal and cardiac muscle and are prime targets for modification of contraction in disorders that affect either the skeletal or heart musculature. We designed and synthesized a number of compounds with structures based on a naturally occurring peptide (A peptides) that modifies the activity of RyRs. In total, 34 compounds belonging to eight different classes were prepared. The compounds were screened for their ability to enhance Ca2+ release from isolated cardiac sarcoplasmic reticulum (SR) vesicles, with 25 displaying enhanced Ca2+ release. Competition studies with the parent peptides indicated that the synthetic compounds act at a competing site. The activity of the most effective of the compounds, BIT 180, was further explored using Ca2+ release from skeletal SR vesicles and contraction in intact skeletal muscle fibers. The compounds did not alter tension in intact fibers, indicating that (as expected) they are not membrane permeable, but importantly, that they are not toxic to the intact cells. Proof in principal that the compounds would be effective in intact muscle fibers if rendered membrane permeable was obtained with a structurally related membrane-permeable scorpion toxin (imperatoxin A), which was found to enhance contraction., (© 2018 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)- Published
- 2018
- Full Text
- View/download PDF
24. Reviewing Hit Discovery Literature for Difficult Targets: Glutathione Transferase Omega-1 as an Example.
- Author
-
Xie Y, Dahlin JL, Oakley AJ, Casarotto MG, Board PG, and Baell JB
- Subjects
- Drug Design, Drug Discovery, Fluorescence Polarization methods, Glutathione Transferase metabolism, Humans, Drug Evaluation, Preclinical methods, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Glutathione Transferase antagonists & inhibitors, Glutathione Transferase chemistry
- Abstract
Early stage drug discovery reporting on relatively new or difficult targets is often associated with insufficient hit triage. Literature reviews of such targets seldom delve into the detail required to critically analyze the associated screening hits reported. Here we take the enzyme glutathione transferase omega-1 (GSTO1-1) as an example of a relatively difficult target and review the associated literature involving small-molecule inhibitors. As part of this process we deliberately pay closer-than-usual attention to assay interference and hit quality aspects. We believe this Perspective will be a useful guide for future development of GSTO1-1 inhibitors, as well serving as a template for future review formats of new or difficult targets.
- Published
- 2018
- Full Text
- View/download PDF
25. Functional and structural characterization of a novel malignant hyperthermia-susceptible variant of DHPR-β 1a subunit (CACNB1).
- Author
-
Perez CF, Eltit JM, Lopez JR, Bodnár D, Dulhunty AF, Aditya S, and Casarotto MG
- Subjects
- Animals, Caffeine pharmacology, Calcium metabolism, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type drug effects, Calcium Channels, L-Type genetics, Cells, Cultured, Dose-Response Relationship, Drug, Homozygote, Humans, Kinetics, Malignant Hyperthermia genetics, Malignant Hyperthermia physiopathology, Mice, Knockout, Mutation, Myoblasts drug effects, Protein Domains, Protein Stability, Structure-Activity Relationship, Calcium Channels, L-Type metabolism, Calcium Signaling drug effects, Malignant Hyperthermia metabolism, Myoblasts metabolism
- Abstract
Malignant hyperthermia (MH) susceptibility has been recently linked to a novel variant of β
1a subunit of the dihydropyridine receptor (DHPR), a channel essential for Ca2+ regulation in skeletal muscle. Here we evaluate the effect of the mutant variant V156A on the structure/function of DHPR β1a subunit and assess its role on Ca2+ metabolism of cultured myotubes. Using differential scanning fluorimetry, we show that mutation V156A causes a significant reduction in thermal stability of the Src homology 3/guanylate kinase core domain of β1a subunit. Expression of the variant subunit in β1 -null mouse myotubes resulted in increased sensitivity to caffeine stimulation. Whole cell patch-clamp analysis of β1a -V156A-expressing myotubes revealed a -2 mV shift in voltage dependence of channel activation, but no changes in Ca2+ conductance, current kinetics, or sarcoplasmic reticulum Ca2+ load were observed. Measurement of resting free Ca2+ and Na+ concentrations shows that both cations were significantly elevated in β1a -V156A-expressing myotubes and that these changes were linked to increased rates of plasmalemmal Ca2+ entry through Na+ /Ca2+ exchanger and/or transient receptor potential canonical channels. Overall, our data show that mutant variant V156A results in instability of protein subdomains of β1a subunit leading to a phenotype of Ca2+ dysregulation that partly resembles that of other MH-linked mutations of DHPR α1S subunit. These data prove that homozygous expression of variant β1a -V156A has the potential to be a pathological variant, although it may require other gene defects to cause a full MH phenotype.- Published
- 2018
- Full Text
- View/download PDF
26. GSTO1-1 plays a pro-inflammatory role in models of inflammation, colitis and obesity.
- Author
-
Menon D, Innes A, Oakley AJ, Dahlstrom JE, Jensen LM, Brüstle A, Tummala P, Rooke M, Casarotto MG, Baell JB, Nguyen N, Xie Y, Cuellar M, Strasser J, Dahlin JL, Walters MA, Burgio G, O'Neill LAJ, and Board PG
- Subjects
- Animals, Carrier Proteins genetics, Colitis drug therapy, Colitis genetics, Glutathione Transferase genetics, Inflammation drug therapy, Inflammation genetics, Male, Mice, Mice, Inbred C57BL, Myeloid Differentiation Factor 88 genetics, Myeloid Differentiation Factor 88 metabolism, Obesity drug therapy, Obesity genetics, Small Molecule Libraries therapeutic use, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 metabolism, Carrier Proteins metabolism, Colitis metabolism, Glutathione Transferase metabolism, Inflammation metabolism, Obesity metabolism
- Abstract
Glutathione transferase Omega 1 (GSTO1-1) is an atypical GST reported to play a pro-inflammatory role in response to LPS. Here we show that genetic knockout of Gsto1 alters the response of mice to three distinct inflammatory disease models. GSTO1-1 deficiency ameliorates the inflammatory response stimulated by LPS and attenuates the inflammatory impact of a high fat diet on glucose tolerance and insulin resistance. In contrast, GSTO1-1 deficient mice show a more severe inflammatory response and increased escape of bacteria from the colon into the lymphatic system in a dextran sodium sulfate mediated model of inflammatory bowel disease. These responses are similar to those of TLR4 and MyD88 deficient mice in these models and confirm that GSTO1-1 is critical for a TLR4-like pro-inflammatory response in vivo. In wild-type mice, we show that a small molecule inhibitor that covalently binds in the active site of GSTO1-1 can be used to ameliorate the inflammatory response to LPS. Our findings demonstrate the potential therapeutic utility of GSTO1-1 inhibitors in the modulation of inflammation and suggest their possible application in the treatment of a range of inflammatory conditions.
- Published
- 2017
- Full Text
- View/download PDF
27. Association of FK506 binding proteins with RyR channels - effect of CLIC2 binding on sub-conductance opening and FKBP binding.
- Author
-
Richardson SJ, Steele GA, Gallant EM, Lam A, Schwartz CE, Board PG, Casarotto MG, Beard NA, and Dulhunty AF
- Subjects
- Animals, Ion Channel Gating, Membrane Potentials, Mutation, Missense, Protein Binding, Rabbits, Sheep, Domestic, Chloride Channels metabolism, Ryanodine Receptor Calcium Release Channel metabolism, Tacrolimus Binding Proteins metabolism
- Abstract
Ryanodine receptor (RyR) Ca
2+ channels are central to striated muscle function and influence signalling in neurons and other cell types. Beneficially low RyR activity and maximum conductance opening may be stabilised when RyRs bind to FK506 binding proteins (FKBPs) and destabilised by FKBP dissociation, with submaximal opening during RyR hyperactivity associated with myopathies and neurological disorders. However, the correlation with submaximal opening is debated and quantitative evidence is lacking. Here, we have measured altered FKBP binding to RyRs and submaximal activity with addition of wild-type (WT) CLIC2, an inhibitory RyR ligand, or its H101Q mutant that hyperactivates RyRs, which probably causes cardiac and intellectual abnormalities. The proportion of sub-conductance opening increases with WT and H101Q CLIC2 and is correlated with reduced FKBP-RyR association. The sub-conductance opening reduces RyR currents in the presence of WT CLIC2. In contrast, sub-conductance openings contribute to excess RyR 'leak' with H101Q CLIC2. There are significant FKBP and RyR isoform-specific actions of CLIC2, rapamycin and FK506 on FKBP-RyR association. The results show that FKBPs do influence RyR gating and would contribute to excess Ca2+ release in this CLIC2 RyR channelopathy., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2017. Published by The Company of Biologists Ltd.)- Published
- 2017
- Full Text
- View/download PDF
28. Publisher's Note: Association of FK506 binding proteins with RyR channels - effect of CLIC2 binding on sub-conductance opening and FKBP binding. J. Cell Sci. doi: 10.1242/jcs.204461.
- Author
-
Richardson SJ, Steele GA, Gallant EM, Lam A, Schwartz CE, Board PG, Casarotto MG, Beard NA, and Dulhunty AF
- Published
- 2017
- Full Text
- View/download PDF
29. Structural and biophysical analyses of the skeletal dihydropyridine receptor β subunit β 1a reveal critical roles of domain interactions for stability.
- Author
-
Norris NC, Joseph S, Aditya S, Karunasekara Y, Board PG, Dulhunty AF, Oakley AJ, and Casarotto MG
- Subjects
- Allosteric Regulation, Animals, Calcium Channels, L-Type genetics, Calcium Channels, L-Type metabolism, Crystallography, X-Ray, Guanylate Kinases genetics, Guanylate Kinases metabolism, Mice, Protein Structure, Secondary, Signal Transduction, src Homology Domains, Calcium Channels, L-Type chemistry, Guanylate Kinases chemistry
- Abstract
Excitation-contraction (EC) coupling in skeletal muscle requires a physical interaction between the voltage-gated calcium channel dihydropyridine receptor (DHPR) and the ryanodine receptor Ca
2+ release channel. Although the exact molecular mechanism that initiates skeletal EC coupling is unresolved, it is clear that both the α1 and β subunits of DHPR are essential for this process. Here, we employed a series of techniques, including size-exclusion chromatography-multi-angle light scattering, differential scanning fluorimetry, and isothermal calorimetry, to characterize various biophysical properties of the skeletal DHPR β subunit β1a Removal of the intrinsically disordered N and C termini and the hook region of β1a prevented oligomerization, allowing for its structural determination by X-ray crystallography. The structure had a topology similar to that of previously determined β isoforms, which consist of SH3 and guanylate kinase domains. However, transition melting temperatures derived from the differential scanning fluorimetry experiments indicated a significant difference in stability of ∼2-3 °C between the β1a and β2a constructs, and the addition of the DHPR α1s I-II loop (α-interaction domain) peptide stabilized both β isoforms by ∼6-8 °C. Similar to other β isoforms, β1a bound with nanomolar affinity to the α-interaction domain, but binding affinities were influenced by amino acid substitutions in the adjacent SH3 domain. These results suggest that intramolecular interactions between the SH3 and guanylate kinase domains play a role in the stability of β1a while also providing a conduit for allosteric signaling events., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2017
- Full Text
- View/download PDF
30. Physiology and Pharmacology of Ryanodine Receptor Calcium Release Channels.
- Author
-
Dulhunty AF, Board PG, Beard NA, and Casarotto MG
- Subjects
- Animals, Arrhythmias, Cardiac metabolism, Humans, Arrhythmias, Cardiac physiopathology, Muscular Diseases physiopathology, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Ryanodine receptor (RyR) ion channels are essential for skeletal and cardiac muscle function. Their knockout leads to perinatal death from respiratory and cardiac failure. Acquired changes or mutations in the protein cause debilitating skeletal myopathy and cardiac arrhythmia which can be deadly. Knowledge of the pharmacology of RyR channels is central to developing effective and specific treatments of these myopathies. The ion channel is a >2.2MDa homotetamer with distinct structural and functional characteristics giving rise to a myriad of regulatory sites that are potential therapeutic targets. Australian researchers have been intimately involved in the exploration of the proteins since their identification in the mid-1980s. We discuss major aspects of RyR physiology and pharmacology that have been tackled in Australian laboratories. Specific areas of interest include ultrastructural aspects and mechanisms of RyR activation in excitation-contraction (EC) coupling and related pharmacological developments, regulation of RyRs by divalent cations, by associated proteins including the FK506-binding proteins, by redox factors and phosphorylation. We consider adverse effects of anthracycline chemotherapeutic drugs on cardiac RyRs. Phenotypes associated with RyR mutations are discussed with current and developing therapeutic approaches for treating the underlying RyR dysfunction., (© 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
31. Core skeletal muscle ryanodine receptor calcium release complex.
- Author
-
Dulhunty AF, Wei-LaPierre L, Casarotto MG, and Beard NA
- Subjects
- Amino Acid Sequence, Animals, Humans, Muscle Proteins chemistry, Muscle Proteins genetics, Muscle Proteins metabolism, Protein Binding physiology, Protein Structure, Secondary, Ryanodine Receptor Calcium Release Channel chemistry, Ryanodine Receptor Calcium Release Channel genetics, Calcium metabolism, Muscle, Skeletal metabolism, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
The core skeletal muscle ryanodine receptor (RyR1) calcium release complex extends through three compartments of the muscle fibre, linking the extracellular environment through the cytoplasmic junctional gap to the lumen of the internal sarcoplasmic reticulum (SR) calcium store. The protein complex is essential for skeletal excitation-contraction (EC)-coupling and skeletal muscle function. Its importance is highlighted by perinatal death if any one of the EC-coupling components are missing and by myopathies associated with mutation of any of the proteins. The proteins essential for EC-coupling include the DHPR α
1S subunit in the transverse tubule membrane, the DHPR β1a subunit in the cytosol and the RyR1 ion channel in the SR membrane. The other core proteins are triadin and junctin and calsequestrin, associated mainly with SR. These SR proteins are not essential for survival but exert structural and functional influences that modify the gain of EC-coupling and maintain normal muscle function. This review summarises our current knowledge of the individual protein/protein interactions within the core complex and their overall contribution to EC-coupling. We highlight significant areas that provide a continuing challenge for the field. Additional important components of the Ca2+ release complex, such as FKBP12, calmodulin, S100A1 and Stac3 are identified and reviewed elsewhere., (© 2016 John Wiley & Sons Australia, Ltd.)- Published
- 2017
- Full Text
- View/download PDF
32. The GSTM2 C-Terminal Domain Depresses Contractility and Ca2+ Transients in Neonatal Rat Ventricular Cardiomyocytes.
- Author
-
Hewawasam RP, Liu D, Casarotto MG, Board PG, and Dulhunty AF
- Subjects
- Actinin, Animals, Animals, Newborn, Electric Stimulation, Glutathione Transferase chemistry, Models, Molecular, Protein Conformation, Rats, Calcium Signaling, Glutathione Transferase metabolism, Heart Ventricles metabolism, Myocardial Contraction, Myocytes, Cardiac metabolism, Protein Interaction Domains and Motifs
- Abstract
The cardiac ryanodine receptor (RyR2) is an intracellular ion channel that regulates Ca2+ release from the sarcoplasmic reticulum (SR) during excitation-contraction coupling in the heart. The glutathione transferases (GSTs) are a family of phase II detoxification enzymes with additional functions including the selective inhibition of RyR2, with therapeutic implications. The C-terminal half of GSTM2 (GSTM2C) is essential for RyR2 inhibition, and mutations F157A and Y160A within GSTM2C prevent the inhibitory action. Our objective in this investigation was to determine whether GSTM2C can enter cultured rat neonatal ventricular cardiomyocytes and influence contractility. We show that oregon green-tagged GSTM2C (at 1 μM) is internalized into the myocytes and it reduces spontaneous contraction frequency and myocyte shortening. Field stimulation of myocytes evoked contraction in the same percentage of myocytes treated either with media alone or media plus 15 μM GSTM2C. Myocyte shortening during contraction was significantly reduced by exposure to 15 μM GSTM2C, but not 5 and 10 μM GSTM2C and was unaffected by exposure to 15 μM of the mutants Y160A or F157A. The amplitude of the Ca2+ transient in the 15 μM GSTM2C - treated myocytes was significantly decreased, the rise time was significantly longer and the decay time was significantly shorter than in control myocytes. The Ca2+ transient was not altered by exposure to Y160A or F157A. The results are consistent with GSTM2C entering the myocytes and inhibiting RyR2, in a manner that indicates a possible therapeutic potential for treatment of arrhythmia in the neonatal heart., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
33. Nuclear PKC-θ facilitates rapid transcriptional responses in human memory CD4+ T cells through p65 and H2B phosphorylation.
- Author
-
Li J, Hardy K, Phetsouphanh C, Tu WJ, Sutcliffe EL, McCuaig R, Sutton CR, Zafar A, Munier CM, Zaunders JJ, Xu Y, Theodoratos A, Tan A, Lim PS, Knaute T, Masch A, Zerweck J, Brezar V, Milburn PJ, Dunn J, Casarotto MG, Turner SJ, Seddiki N, Kelleher AD, and Rao S
- Subjects
- Amino Acid Sequence, Chromatin metabolism, Gene Expression Regulation, Histones chemistry, Humans, Jurkat Cells, Phosphorylation, Phosphoserine metabolism, Protein Kinase C-theta, Signal Transduction, CD4-Positive T-Lymphocytes metabolism, Cell Nucleus enzymology, Histones metabolism, Immunologic Memory genetics, Isoenzymes metabolism, Protein Kinase C metabolism, Transcription Factor RelA metabolism, Transcription, Genetic
- Abstract
Memory T cells are characterized by their rapid transcriptional programs upon re-stimulation. This transcriptional memory response is facilitated by permissive chromatin, but exactly how the permissive epigenetic landscape in memory T cells integrates incoming stimulatory signals remains poorly understood. By genome-wide ChIP-sequencing ex vivo human CD4(+) T cells, here, we show that the signaling enzyme, protein kinase C theta (PKC-θ) directly relays stimulatory signals to chromatin by binding to transcriptional-memory-responsive genes to induce transcriptional activation. Flanked by permissive histone modifications, these PKC-enriched regions are significantly enriched with NF-κB motifs in ex vivo bulk and vaccinia-responsive human memory CD4(+) T cells. Within the nucleus, PKC-θ catalytic activity maintains the Ser536 phosphorylation on the p65 subunit of NF-κB (also known as RelA) and can directly influence chromatin accessibility at transcriptional memory genes by regulating H2B deposition through Ser32 phosphorylation. Furthermore, using a cytoplasm-restricted PKC-θ mutant, we highlight that chromatin-anchored PKC-θ integrates activating signals at the chromatin template to elicit transcriptional memory responses in human memory T cells., (© 2016. Published by The Company of Biologists Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
34. Probing interactions of Vpu from HIV-1 with amiloride-based compounds.
- Author
-
Rosenberg MR, Weaver LM, and Casarotto MG
- Subjects
- Amiloride pharmacology, Amino Acid Sequence drug effects, HIV-1 pathogenicity, Human Immunodeficiency Virus Proteins metabolism, Humans, Ion Channels chemistry, Ion Channels metabolism, Mutation, Surface Plasmon Resonance, Viral Regulatory and Accessory Proteins metabolism, Virus Replication drug effects, HIV-1 chemistry, Human Immunodeficiency Virus Proteins chemistry, Liposomes chemistry, Viral Regulatory and Accessory Proteins chemistry
- Abstract
Viral ion channels or viroporins are short membrane proteins that participate in wide-ranging functions including virus replication and entry, assembly, and virus release. One such viroporin is the 81 amino acid residue Vpu protein derived from HIV-1. This protein consists of one transmembrane (TM) and two cytoplasmic helical domains, the former of which oligomerises to form cation-selective ion channels. In this study, we investigate the binding properties of amiloride compounds to Vpu embedded into liposomes using surface plasmon resonance (SPR). We explore the Vpu ion channel inhibitor, hexamethylene amiloride (HMA), as a molecular tool to examine the potential interactive role of key TM residues, Trp23, Ser24, and Glu29, in terms of positioning of these residues on the channel pore and the orientation of its constituent helices. The study provides experimental support that a direct interaction between Ser24 and HMA occurs and that this residue is most likely located in the channel pore. Mutation of Trp23 does not impact HMA affinity suggesting no direct involvement in binding and that this residue is lipid facing. These findings indicate that small molecules such as amilorides are capable of specifically interacting with Vpu ion channels. Although a correlation between ion channel and functional activity cannot be dismissed, alternative mechanisms involving protein-protein interactions may play an important role in the efficacy of these compounds., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. Glutathione transferase M2 variants inhibit ryanodine receptor function in adult mouse cardiomyocytes.
- Author
-
Samarasinghe K, Liu D, Tummala P, Cappello J, Pace SM, Arnolda L, Casarotto MG, Dulhunty AF, and Board PG
- Subjects
- Animals, Caffeine pharmacology, Cells, Cultured, Circular Dichroism, Escherichia coli genetics, Male, Mice, Inbred C57BL, Mutagenesis, Site-Directed, Myocytes, Cardiac metabolism, Peptide Fragments genetics, Two-Hybrid System Techniques, Calcium Channel Blockers pharmacology, Glutathione Transferase genetics, Myocytes, Cardiac drug effects, Peptide Fragments pharmacology, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Release of Ca(2+) from the sarcoplasmic reticulum (SR) through the cardiac ryanodine receptor (RyR2) is an essential step in cardiac excitation-contraction coupling. Excess Ca(2+) release due to overactive RyR2 can cause arrhythmia that can lead to cardiac arrest. Fragments derived from the carboxy-terminal domain of human glutathione transferase M2 (GSTM2C) specifically inhibit RyR2 activity. Our aim was to further improve this inhibition by mutagenesis and to assess the therapeutic potential of GSTM2C based peptides to treat Ca(2+) release-based arrhythmia. We generated several mutant variants of the C-terminal fragment GSTM2C H5-8 and from those mutant proteins we identified two (RM13 and SM2) that exhibited significantly greater inhibition of cardiac SR Ca(2+) release and single RyR2 channel activity. Flow cytometry analysis showed that these two mutant proteins as well as GSTM2C H5-8 are taken up by isolated adult mouse cardiomyocytes without the aid of any additional compounds, Ca(2+) imaging and isolated cell contraction measurements revealed that GSTM2C H5-8, SM2 and RM13 reduce the SR Ca(2+) release rate and the fractional shortening of adult mouse cardiomyocytes, while importantly increasing the rate of Ca(2+) removal from the sarcoplasm. These observations indicate that peptides derived from GSTM2C inhibit RyR2 at a cellular level and thus they may provide the basis for a novel therapeutic agent to treat arrhythmia and heart attack., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. Regions of ryanodine receptors that influence activation by the dihydropyridine receptor β1a subunit.
- Author
-
Rebbeck RT, Willemse H, Groom L, Casarotto MG, Board PG, Beard NA, Dirksen RT, and Dulhunty AF
- Abstract
Background: Although excitation-contraction (EC) coupling in skeletal muscle relies on physical activation of the skeletal ryanodine receptor (RyR1) Ca(2+) release channel by dihydropyridine receptors (DHPRs), the activation pathway between the DHPR and RyR1 remains unknown. However, the pathway includes the DHPR β1a subunit which is integral to EC coupling and activates RyR1. In this manuscript, we explore the isoform specificity of β1a activation of RyRs and the β1a binding site on RyR1., Methods: We used lipid bilayers to measure single channel currents and whole cell patch clamp to measure L-type Ca(2+) currents and Ca(2+) transients in myotubes., Results: We demonstrate that both skeletal RyR1 and cardiac RyR2 channels in phospholipid bilayers are activated by 10-100 nM of the β1a subunit. Activation of RyR2 by 10 nM β1a was less than that of RyR1, suggesting a reduced affinity of RyR2 for β1a. A reduction in activation was also observed when 10 nM β1a was added to the alternatively spliced (ASI(-)) isoform of RyR1, which lacks ASI residues (A3481-Q3485). It is notable that the equivalent region of RyR2 also lacks four of five ASI residues, suggesting that the absence of these residues may contribute to the reduced 10 nM β1a activation observed for both RyR2 and ASI(-)RyR1 compared to ASI(+)RyR1. We also investigated the influence of a polybasic motif (PBM) of RyR1 (K3495KKRRDGR3502) that is located immediately downstream from the ASI residues and has been implicated in EC coupling. We confirmed that neutralizing the basic residues in the PBM (RyR1 K-Q) results in an ~50 % reduction in Ca(2+) transient amplitude following expression in RyR1-null (dyspedic) myotubes and that the PBM is also required for β1a subunit activation of RyR1 channels in lipid bilayers. These results suggest that the removal of β1a subunit interaction with the PBM in RyR1 could contribute directly to ~50 % of the Ca(2+) release generated during skeletal EC coupling., Conclusions: We conclude that the β1a subunit likely binds to a region that is largely conserved in RyR1 and RyR2 and that this region is influenced by the presence of the ASI residues and the PBM in RyR1.
- Published
- 2015
- Full Text
- View/download PDF
37. Identification of a pathogenic variant in TREX1 in early-onset cerebral systemic lupus erythematosus by Whole-exome sequencing.
- Author
-
Ellyard JI, Jerjen R, Martin JL, Lee AY, Field MA, Jiang SH, Cappello J, Naumann SK, Andrews TD, Scott HS, Casarotto MG, Goodnow CC, Chaitow J, Pascual V, Hertzog P, Alexander SI, Cook MC, and Vinuesa CG
- Subjects
- Child, Preschool, Female, Humans, Pedigree, Exodeoxyribonucleases genetics, Exome genetics, Homozygote, Interferon-alpha analysis, Lupus Vasculitis, Central Nervous System genetics, Phosphoproteins genetics
- Abstract
Objective. Systemic lupus erythematosus (SLE) isa chronic and heterogeneous autoimmune disease. Both twin and sibling studies indicate a strong genetic contribution to lupus, but in the majority of cases the pathogenic variant remains to be identified. The genetic contribution to disease is likely to be greatest in cases with early onset and severe phenotypes. Whole-exome sequencing now offers the possibility of identifying rare alleles responsible for disease in such cases. This study was undertaken to identify genetic causes of SLE using whole-exome sequencing.Methods. We performed whole-exome sequencing in a 4-year-old girl with early-onset SLE and conducted biochemical analysis of the putative defect.Results. Whole-exome sequencing in a 4-year-old girl with cerebral lupus identified a rare, homozygous mutation in the three prime repair exonuclease 1 gene(TREX1) that was predicted to be highly deleterious.The TREX1 R97H mutant protein had a 20-fold reduction in exonuclease activity and was associated with an elevated interferon-alpha signature in the patient.The discovery and characterization of a pathogenic TREX1 variant in our proband has therapeutic implications.The patient is now a candidate for therapy. Conclusion. Our study is the first to demonstrate that whole-exome sequencing can be used to identify rare or novel deleterious variants as genetic causes of SLE and, through a personalized approach, improve therapeutic options.
- Published
- 2014
- Full Text
- View/download PDF
38. Skeletal muscle excitation-contraction coupling: who are the dancing partners?
- Author
-
Rebbeck RT, Karunasekara Y, Board PG, Beard NA, Casarotto MG, and Dulhunty AF
- Subjects
- Animals, Excitation Contraction Coupling, Humans, Muscle Proteins metabolism, Muscle, Skeletal metabolism, Sarcoplasmic Reticulum metabolism, Muscle, Skeletal physiology
- Abstract
There is an overwhelming body of work supporting the idea that excitation-contraction coupling in skeletal muscle depends on a physical interaction between the skeletal muscle isoform of the dihydropyridine receptor L-type Ca(2+) channel and the skeletal isoform of the ryanodine receptor Ca(2+) release channel. A general assumption is that this physical interaction is between "critical" residues that have been identified in the II-III loop of the dihydropyridine receptor alpha subunit and the ryanodine receptor. However, despite extensive searches, the complementary "critical" residues in the ryanodine receptor have not been identified. This raises the possibility that the coupling proceeds either through other subunits of the dihydropyridine receptor and/or other co-proteins within the large RyR1 protein complex. There have been some remarkable advances in recent years in identifying proteins in the RyR complex that impact on the coupling process, and these are considered in this review. A major candidate for a role in the coupling mechanism is the beta subunit of the dihydropyridine receptor, because specific residues in both the beta subunit and ryanodine receptor have been identified that facilitate an interaction between the two proteins and these also impact on excitation-contraction coupling. This role of beta subunit remains to be fully investigated as well as the degree to which it may complement any other direct or indirect voltage-dependent coupling interactions between the DHPR alpha II-III loop and the ryanodine receptor., (Copyright © 2014. Published by Elsevier Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
39. An α-helical C-terminal tail segment of the skeletal L-type Ca2+ channel β1a subunit activates ryanodine receptor type 1 via a hydrophobic surface.
- Author
-
Karunasekara Y, Rebbeck RT, Weaver LM, Board PG, Dulhunty AF, and Casarotto MG
- Subjects
- Amino Acid Sequence, Amino Acids chemistry, Amino Acids genetics, Amino Acids metabolism, Animals, Binding Sites genetics, Calcium Channels, L-Type genetics, Excitation Contraction Coupling, Hydrophobic and Hydrophilic Interactions, Mice, Models, Molecular, Molecular Sequence Data, Muscle, Skeletal metabolism, Muscle, Skeletal physiology, Mutation, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Rabbits, Ryanodine Receptor Calcium Release Channel genetics, Sequence Homology, Amino Acid, Surface Properties, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type metabolism, Ryanodine Receptor Calcium Release Channel chemistry, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Excitation-contraction (EC) coupling in skeletal muscle depends on protein interactions between the transverse tubule dihydropyridine receptor (DHPR) voltage sensor and intracellular ryanodine receptor (RyR1) calcium release channel. We present novel data showing that the C-terminal 35 residues of the β(1a) subunit adopt a nascent α-helix in which 3 hydrophobic residues align to form a hydrophobic surface that binds to RyR1 isolated from rabbit skeletal muscle. Mutation of the hydrophobic residues (L496, L500, W503) in peptide β(1a)V490-M524, corresponding to the C-terminal 35 residues of β(1a), reduced peptide binding to RyR1 to 15.2 ± 7.1% and prevented the 2.9 ± 0.2-fold activation of RyR1 by 10 nM wild-type peptide. An upstream hydrophobic heptad repeat implicated in β(1a) binding to RyR1 does not contribute to RyR1 activation. Wild-type β(1a)A474-A508 peptide (10 nM), containing heptad repeat and hydrophobic surface residues, increased RyR1 activity by 2.3 ± 0.2- and 2.2 ± 0.3-fold after mutation of the heptad repeat residues. We conclude that specific hydrophobic surface residues in the 35 residue β(1a) C-terminus bind to RyR1 and increase channel activity in lipid bilayers and thus may support skeletal EC coupling.
- Published
- 2012
- Full Text
- View/download PDF
40. Design and synthesis of pinanamine derivatives as anti-influenza A M2 ion channel inhibitors.
- Author
-
Zhao X, Jie Y, Rosenberg MR, Wan J, Zeng S, Cui W, Xiao Y, Li Z, Tu Z, Casarotto MG, and Hu W
- Subjects
- Animals, Antiviral Agents chemical synthesis, Antiviral Agents chemistry, Cell Line, Humans, Microbial Sensitivity Tests, Molecular Structure, Viral Plaque Assay, Antiviral Agents isolation & purification, Antiviral Agents pharmacology, Influenza A virus drug effects, Ion Channels antagonists & inhibitors, Viral Matrix Proteins antagonists & inhibitors
- Abstract
The adamantanes are a class of anti-influenza drugs that inhibit the M2 ion channel of the influenza A virus. However recently, the clinical effectiveness of these drugs has been called into question due to the emergence of adamantane-insensitive A/M2 mutants. Although we previously reported (1R,2R,3R,5S)-3-pinanamine 3 as a novel inhibitor of the wild type influenza A virus M2 protein (WT A/M2), limited inhibition was found for adamantane-resistant M2 mutants. In this study, we explored whether newly synthesized pinanamine derivatives were capable of inhibiting WT A/M2 and selected adamantane-resistant M2 mutants. Several imidazole and guanazole derivatives of pinanamine were found to inhibit WT A/M2 to a comparable degree as amantadine and one of these compounds 12 exhibits weak inhibition of A/M2-S31N mutant and it is marginally more effective in inhibiting S31NM2 than amantadine. This study provides a new insight into the structural nature of drugs required to inhibit WT A/M2 and its mutants., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
41. Selective modulation of different GABAA receptor isoforms by diazepam and etomidate in hippocampal neurons.
- Author
-
Seymour VA, Curmi JP, Howitt SM, Casarotto MG, Laver DR, and Tierney ML
- Subjects
- Amino Acid Sequence, Animals, Electric Conductivity, Female, Hydrophobic and Hydrophilic Interactions, Male, Models, Molecular, Molecular Sequence Data, Neurons cytology, Permeability drug effects, Protein Isoforms chemistry, Protein Isoforms metabolism, Protein Structure, Secondary, Rats, Rats, Wistar, Receptors, GABA-A chemistry, Diazepam pharmacology, Etomidate pharmacology, Hippocampus cytology, Neurons drug effects, Neurons metabolism, Receptors, GABA-A metabolism
- Abstract
Diazepam modulation of native γ2-containing GABA(A) (γGABA(A)) receptors increases channel conductance by facilitating protein interactions involving the γ2-subunit amphipathic (MA) region, which is found in the cytoplasmic loop between transmembrane domains 3 and 4 (Everitt et al., 2009). However, many drugs, predicted to act on different GABA(A) receptor subtypes, increase channel conductance leading us to hypothesize that conductance variation in GABA(A) receptors may be a general property, mediated by protein interactions involving the cytoplasmic MA stretch of amino acids. In this study we have tested this hypothesis by potentiating extrasynaptic GABA(A) currents with etomidate and examining the ability of peptides mimicking either the γ2- or δ-subunit MA region to affect conductance. In inside-out hippocampal patches from newborn rats the general anesthetic etomidate potentiated GABA currents, producing either an increase in open probability and single-channel conductance or an increase in open probability, as described previously (Seymour et al., 2009). In patches displaying high conductance channels application of a δ((392-422)) MA peptide, but not a scrambled version or the equivalent γ2((381-403)) MA peptide, reduced the potentiating effects of etomidate, significantly reducing single-channel conductance. In contrast, when GABA currents were potentiated by the γ2-specific drug diazepam the δ MA peptide had no effect. These data reveal that diazepam and etomidate potentiate different extrasynaptic GABA(A) receptor subtypes but both drugs modulate conductance similarly. One interpretation of the data is that these drugs elicit potentiation through protein interactions and that the MA peptides compete with these interactions to disrupt this process., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
42. Chromatinized Protein Kinase C-θ: Can It Escape the Clutches of NF-κB?
- Author
-
Sutcliffe EL, Li J, Zafar A, Hardy K, Ghildyal R, McCuaig R, Norris NC, Lim PS, Milburn PJ, Casarotto MG, Denyer G, and Rao S
- Abstract
We recently provided the first description of a nuclear mechanism used by Protein Kinase C-theta (PKC-θ) to mediate T cell gene expression. In this mode, PKC-θ tethers to chromatin to form an active nuclear complex by interacting with proteins including RNA polymerase II, the histone kinase MSK-1, the demethylase LSD1, and the adaptor molecule 14-3-3ζ at regulatory regions of inducible immune response genes. Moreover, our genome-wide analysis identified many novel PKC-θ target genes and microRNAs implicated in T cell development, differentiation, apoptosis, and proliferation. We have expanded our ChIP-on-chip analysis and have now identified a transcription factor motif containing NF-κB binding sites that may facilitate recruitment of PKC-θ to chromatin at coding genes. Furthermore, NF-κB association with chromatin appears to be a prerequisite for the assembly of the PKC-θ active complex. In contrast, a distinct NF-κB-containing module appears to operate at PKC-θ targeted microRNA genes, and here NF-κB negatively regulates microRNA gene transcription. Our efforts are also focusing on distinguishing between the nuclear and cytoplasmic functions of PKCs to ascertain how these kinases may synergize their roles as both cytoplasmic signaling proteins and their functions on the chromatin template, together enabling rapid induction of eukaryotic genes. We have identified an alternative sequence within PKC-θ that appears to be important for nuclear translocation of this kinase. Understanding the molecular mechanisms used by signal transduction kinases to elicit specific and distinct transcriptional programs in T cells will enable scientists to refine current therapeutic strategies for autoimmune diseases and cancer.
- Published
- 2012
- Full Text
- View/download PDF
43. The inhibitory glutathione transferase M2-2 binding site is located in divergent region 3 of the cardiac ryanodine receptor.
- Author
-
Liu D, Hewawasam R, Karunasekara Y, Casarotto MG, Dulhunty AF, and Board PG
- Subjects
- Amino Acid Sequence, Binding Sites, Calcium Signaling, Cloning, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, Protein Binding, Ryanodine Receptor Calcium Release Channel genetics, Sarcoplasmic Reticulum metabolism, Tryptophan chemistry, Tryptophan metabolism, Two-Hybrid System Techniques, Glutathione Transferase metabolism, Ryanodine Receptor Calcium Release Channel chemistry, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
The muscle-specific glutathione transferase GSTM2-2 modulates the activity of ryanodine receptor (RyR) calcium release channels: it inhibits the activity of cardiac RyR (RyR2) channels with high affinity and activates skeletal RyR (RyR1) channels with low affinity. The C terminal domain of GSTM2-2 (GSTM2C) alone physically binds to RyR2 and inhibits its activity, but it does not bind to RyR1. We have now used yeast two-hybrid analysis, chemical cross-linking, intrinsic tryptophan fluorescence and Ca(2+) release studies to determine that the binding site for GSTM2C is in divergent region 3 (D3) of RyR2. The D3 region encompasses residues 1855-1890 in RyR2. Specific mutagenesis shows the binding primarily involves electrostatic interactions with residues K1875, K1886, R1887 and K1889, all residues that are present in RyR2, but not in RyR1. The significant sequence differences between the D3 regions of RyR2 and RyR1 explain why GSTM2-2 specifically inhibits RyR2. This specific inhibition of RyR2 could modulate Ca cycling and be useful for the treatment of heart failure. RyR2 inhibition during diastole may improve filling of the SR with Ca(2+) and improve contractility., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
44. Cyclization of the intrinsically disordered α1S dihydropyridine receptor II-III loop enhances secondary structure and in vitro function.
- Author
-
Tae HS, Cui Y, Karunasekara Y, Board PG, Dulhunty AF, and Casarotto MG
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Calcium Channels, L-Type genetics, Cyclization, DnaB Helicases chemistry, DnaB Helicases metabolism, Inteins genetics, Ion Channel Gating, Lipid Bilayers metabolism, Molecular Sequence Data, Muscle Contraction genetics, Muscle, Skeletal metabolism, Muscle, Skeletal physiology, Protein Splicing, Protein Structure, Secondary, Protein Structure, Tertiary, Protein Subunits chemistry, Protein Subunits genetics, Protein Subunits metabolism, Rabbits, Ryanodine Receptor Calcium Release Channel metabolism, Substrate Specificity, Synechocystis enzymology, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type metabolism, Protein Engineering methods
- Abstract
A key component of excitation contraction (EC) coupling in skeletal muscle is the cytoplasmic linker (II-III loop) between the second and third transmembrane repeats of the α(1S) subunit of the dihydropyridine receptor (DHPR). The II-III loop has been previously examined in vitro using a linear II-III loop with unrestrained N- and C-terminal ends. To better reproduce the loop structure in its native environment (tethered to the DHPR transmembrane domains), we have joined the N and C termini using intein-mediated technology. Circular dichroism and NMR spectroscopy revealed a structural shift in the cyclized loop toward a protein with increased α-helical and β-strand structure in a region of the loop implicated in its in vitro function and also in a critical region for EC coupling. The affinity of binding of the II-III loop binding to the SPRY2 domain of the skeletal ryanodine receptor (RyR1) increased 4-fold, and its ability to activate RyR1 channels in lipid bilayers was enhanced 3-fold by cyclization. These functional changes were predicted consequences of the structural enhancement. We suggest that tethering the N and C termini stabilized secondary structural elements in the DHPR II-III loop and may reflect structural and dynamic characteristics of the loop that are inherent in EC coupling.
- Published
- 2011
- Full Text
- View/download PDF
45. The ryanodine receptor: a pivotal Ca2+ regulatory protein and potential therapeutic drug target.
- Author
-
Dulhunty AF, Casarotto MG, and Beard NA
- Subjects
- Animals, Arrhythmias, Cardiac drug therapy, Arrhythmias, Cardiac genetics, Arrhythmias, Cardiac metabolism, Calcium Signaling, Disease Models, Animal, Heart Diseases drug therapy, Heart Diseases genetics, Heart Failure drug therapy, Heart Failure metabolism, Humans, Malignant Hyperthermia drug therapy, Malignant Hyperthermia genetics, Mutation, Myopathy, Central Core drug therapy, Myopathy, Central Core genetics, Calcium metabolism, Heart Diseases metabolism, Malignant Hyperthermia metabolism, Molecular Targeted Therapy, Myopathy, Central Core metabolism, Ryanodine Receptor Calcium Release Channel genetics, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
The ryanodine receptor (RyR) calcium release channel is an essential intracellular ion channel that is central to Ca(2+) signaling and contraction in the heart and skeletal muscle. The rapid release of Ca(2+) from the internal sarcoplasmic reticulum Ca(2+) stores through the RyR during excitation-contraction coupling is facilitated by the unique arrangement of the surface and sarcoplasmic reticulum membrane systems. Debilitating and sometimes fatal skeletal and cardiomyopathies result from changes in RyR activity that disrupt normal Ca(2+) signaling. Such changes can be caused by point mutations in many different regions of the RyR protein or acquired as a result of stress associated with exercise, heart failure, age or drugs. In general, both inherited and acquired changes include an increase in RyR channel activity. Because of its central function, the RyR is a potential therapeutic target for the inherited disorders and many of the acquired disorders. The RyR is currently used as a therapeutic target in malignant hyperthermia where dantrolene is effective and to relieve ventricular arrhythmia, with the use of JTV519 and flecainide. These drugs show that the RyR is a valid therapeutic target, but have side effects that prevent their chronic use. Thus there is an urgent need for the development of skeletal and cardiac specific drugs to treat these diverse muscle disorders. In this review, we discuss the mutations that cause skeletal myopathies and cardiac arrhythmias and how these mutations pinpoint residues within the RyR protein that are functionally significant and might be developed as targets for therapeutic drugs.
- Published
- 2011
- Full Text
- View/download PDF
46. Regulation of the cardiac muscle ryanodine receptor by glutathione transferases.
- Author
-
Dulhunty AF, Hewawasam R, Liu D, Casarotto MG, and Board PG
- Subjects
- Animals, Binding Sites, Cytosol enzymology, Cytosol metabolism, Glutathione Transferase chemistry, Glutathione Transferase metabolism, Humans, Isoenzymes, Models, Molecular, Myocardium enzymology, Oxidation-Reduction, Protein Conformation, Glutathione Transferase physiology, Myocardium metabolism, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Glutathione transferases (GSTs) are generally recognized for their role in phase II detoxification reactions. However, it is becoming increasingly apparent that members of the GST family also have a diverse range of other functions that are, in general, unrelated to detoxification. One such action is a specific inhibition of the cardiac isoform of the ryanodine receptor (RyR2) intracellular Ca(2+) release channel. In this review, we compare functional and physical interactions between members of the GST family, including GSTO1-1, GSTA1-1, and GSTM2-2, with RyR2 and with the skeletal isoform of the ryanodine receptor (RyR1). The active part of the muscle-specific GSTM2-2 is localized to its nonenzymatic C-terminal α-helical bundle, centered around α-helix 6. The GSTM2-2 binding site is in divergent region 3 (DR3 region) of RyR2. The sequence differences between the DR3 regions of RyR1 and RyR2 explain the specificity of the GSTs for one isoform of the protein. GSTM2-2 is one of the few known endogenous inhibitors of the cardiac RyR and is likely to be important in maintaining low RyR2 activity during diastole. We discuss interactions between a nonenzymatic member of the GST structural family, the CLIC-2 (type 2 chloride intracellular channel) protein, which inhibits both RyR1 and RyR2. The possibility that the GST and CLIC2 proteins bind to different sites on the RyR, and that different structures within the GST and CLIC proteins bind to RyR channels, is discussed. We conclude that the C-terminal part of GSTM2-2 may provide the basis of a therapeutic compound for use in cardiac disorders.
- Published
- 2011
- Full Text
- View/download PDF
47. A structural basis for cellular uptake of GST-fold proteins.
- Author
-
Morris MJ, Liu D, Weaver LM, Board PG, and Casarotto MG
- Subjects
- Animals, Cell Line, Circular Dichroism, Flow Cytometry, Fluorometry, Glutathione Transferase genetics, Humans, Mice, Microscopy, Confocal, Protein Structure, Secondary, Protein Transport genetics, Protein Transport physiology, Recombinant Proteins genetics, Glutathione Transferase metabolism, Recombinant Proteins metabolism
- Abstract
It has recently emerged that glutathione transferase enzymes (GSTs) and other structurally related molecules can be translocated from the external medium into many different cell types. In this study we aim to explore in detail, the structural features that govern cell translocation and by dissecting the human GST enzyme GSTM2-2 we quantatively demonstrate that the α-helical C-terminal domain (GST-C) is responsible for this property. Attempts to further examine the constituent helices within GST-C resulted in a reduction in cell translocation efficiency, indicating that the intrinsic GST-C domain structure is necessary for maximal cell translocation capacity. In particular, it was noted that the α-6 helix of GST-C plays a stabilising role in the fold of this domain. By destabilising the conformation of GST-C, an increase in cell translocation efficiency of up to ∼2-fold was observed. The structural stability profiles of these protein constructs have been investigated by circular dichroism and differential scanning fluorimetry measurements and found to impact upon their cell translocation efficiency. These experiments suggest that the globular, helical domain in the 'GST-fold' structural motif plays a role in influencing cellular uptake, and that changes that affect the conformational stability of GST-C can significantly influence cell translocation efficiency.
- Published
- 2011
- Full Text
- View/download PDF
48. The elusive role of the SPRY2 domain in RyR1.
- Author
-
Tae H, Wei L, Willemse H, Mirza S, Gallant EM, Board PG, Dirksen RT, Casarotto MG, and Dulhunty A
- Subjects
- Alternative Splicing, Animals, Cytoplasm metabolism, Humans, Kinetics, Membrane Proteins, Muscle Fibers, Skeletal metabolism, Muscle, Skeletal metabolism, Protein Binding, Protein Structure, Tertiary, Rabbits, Sarcoplasmic Reticulum metabolism, gamma-Glutamylcyclotransferase metabolism, Dihydropteridine Reductase genetics, Intracellular Signaling Peptides and Proteins chemistry, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
The second of three SPRY domains (SPRY2, S1085 -V1208) located in the skeletal muscle ryanodine receptor (RyR1) is contained within regions of RyR1 that influence EC coupling and bind to imperatoxin A, a toxin probe of RyR1 channel gating. We examined the binding of the F loop (P1107-A1121) in SPRY2 to the ASI/basic region in RyR1 (T3471-G3500, containing both alternatively spliced (ASI) residues and neighboring basic amino acids). We then investigated the possible influence of this interaction on excitation contraction (EC) coupling. A peptide with the F loop sequence and an antibody to the SPRY2 domain each enhanced RyR1 activity at low concentrations and inhibited at higher concentrations. A peptide containing the ASI/basic sequence bound to SPRY2 and binding decreased ~10-fold following mutation or structural disruption of the basic residues. Binding was abolished by mutation of three critical acidic F loop residues. Together these results suggest that the ASI/basic and SPRY2 domains interact in an F loop regulatory module. Although a region that includes the SPRY2 domain influences EC coupling, as does the ASI/basic region, Ca2+ release during ligand- and depolarization-induced RyR1 activation were not altered by mutation of the three critical F loop residues following expression of mutant RyR1 in RyR1-null myotubes. Therefore the electrostatic regulatory interaction between the SPRY2 F loop residues (that bind to imperatoxin A) and the ASI/basic residues of RyR1 does not influence bi-directional DHPR-RyR1 signaling during skeletal EC coupling, possibly because the interaction is interrupted by the influence of factors present in intact muscle cells.
- Published
- 2011
- Full Text
- View/download PDF
49. The β(1a) subunit of the skeletal DHPR binds to skeletal RyR1 and activates the channel via its 35-residue C-terminal tail.
- Author
-
Rebbeck RT, Karunasekara Y, Gallant EM, Board PG, Beard NA, Casarotto MG, and Dulhunty AF
- Subjects
- Amino Acid Sequence, Animals, Calcium pharmacology, Molecular Sequence Data, Muscle, Skeletal drug effects, Protein Binding drug effects, Protein Stability drug effects, Rabbits, Ryanodine metabolism, Ryanodine Receptor Calcium Release Channel chemistry, Structure-Activity Relationship, Calcium Channels, L-Type chemistry, Calcium Channels, L-Type metabolism, Ion Channel Gating drug effects, Muscle, Skeletal metabolism, Protein Subunits metabolism, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Although it has been suggested that the C-terminal tail of the β(1a) subunit of the skeletal dihyropyridine receptor (DHPR) may contribute to voltage-activated Ca(2+) release in skeletal muscle by interacting with the skeletal ryanodine receptor (RyR1), a direct functional interaction between the two proteins has not been demonstrated previously. Such an interaction is reported here. A peptide with the sequence of the C-terminal 35 residues of β(1a) bound to RyR1 in affinity chromatography. The full-length β(1a) subunit and the C-terminal peptide increased [(3)H]ryanodine binding and RyR1 channel activity with an AC(50) of 450-600 pM under optimal conditions. The effect of the peptide was dependent on cytoplasmic Ca(2+), ATP, and Mg(2+) concentrations. There was no effect of the peptide when channel activity was very low as a result of Mg(2+) inhibition or addition of 100 nM Ca(2+) (without ATP). Maximum increases were seen with 1-10 μM Ca(2+), in the absence of Mg(2+) inhibition. A control peptide with the C-terminal 35 residues in a scrambled sequence did not bind to RyR1 or alter [(3)H]ryanodine binding or channel activity. This high-affinity in vitro functional interaction between the C-terminal 35 residues of the DHPR β(1a) subunit and RyR1 may support an in vivo function of β(1a) during voltage-activated Ca(2+) release., (Copyright © 2011 Biophysical Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
50. Novel folding and stability defects cause a deficiency of human glutathione transferase omega 1.
- Author
-
Zhou H, Brock J, Casarotto MG, Oakley AJ, and Board PG
- Subjects
- Crystallography, X-Ray, Enzyme Stability genetics, Escherichia coli, Glutathione Transferase genetics, Glutathione Transferase metabolism, Humans, Mutation, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Urea chemistry, Glutathione Transferase chemistry, Glutathione Transferase deficiency, Models, Molecular, Protein Folding
- Abstract
The polymorphic deletion of Glu-155 from human glutathione transferase omega1 (GSTO1-1) occurs in most populations. Although the recombinant ΔGlu-155 enzyme expressed in Escherichia coli is active, the deletion causes a deficiency of the active enzyme in vivo. The crystal structure and the folding/unfolding kinetics of the ΔGlu-155 variant were determined in order to investigate the cause of the rapid loss of the enzyme in human cells. The crystal structure revealed altered packing around the Glu-155 deletion, an increase in the predicted solvent-accessible area and a corresponding reduction in the buried surface area. This increase in solvent accessibility was consistent with an elevated Stern-Volmer constant. The unfolding of both the wild type and ΔGlu-155 enzyme in urea is best described by a three-state model, and there is evidence for the more pronounced population of an intermediate state by the ΔGlu-155 enzymes. Studies using intrinsic fluorescence revealed a free energy change around 14.4 kcal/mol for the wild type compared with around 8.6 kcal/mol for the ΔGlu-155 variant, which indicates a decrease in stability associated with the Glu-155 deletion. Urea induced unfolding of the wild type GSTO1-1 was reversible through an initial fast phase followed by a second slow phase. In contrast, the ΔGlu-155 variant lacks the slow phase, indicating a refolding defect. It is possible that in some conditions in vivo, the increased solvent-accessible area and the low stability of the ΔGlu-155 variant may promote its unfolding, whereas the refolding defect limits its refolding, resulting in GSTO1-1 deficiency.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.