Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Proc Natl Acad Sci U S A ; 117(25): 14139-14149, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32503917

ABSTRACT

Agonist-activated G protein-coupled receptors (GPCRs) must correctly select from hundreds of potential downstream signaling cascades and effectors. To accomplish this, GPCRs first bind to an intermediary signaling protein, such as G protein or arrestin. These intermediaries initiate signaling cascades that promote the activity of different effectors, including several protein kinases. The relative roles of G proteins versus arrestins in initiating and directing signaling is hotly debated, and it remains unclear how the correct final signaling pathway is chosen given the ready availability of protein partners. Here, we begin to deconvolute the process of signal bias from the dopamine D1 receptor (D1R) by exploring factors that promote the activation of ERK1/2 or Src, the kinases that lead to cell growth and proliferation. We found that ERK1/2 activation involves both arrestin and Gαs, while Src activation depends solely on arrestin. Interestingly, we found that the phosphorylation pattern influences both arrestin and Gαs coupling, suggesting an additional way the cells regulate G protein signaling. The phosphorylation sites in the D1R intracellular loop 3 are particularly important for directing the binding of G protein versus arrestin and for selecting between the activation of ERK1/2 and Src. Collectively, these studies correlate functional outcomes with a physical basis for signaling bias and provide fundamental information on how GPCR signaling is directed.


Subject(s)
Receptors, Dopamine D1/metabolism , Signal Transduction , Arrestin/metabolism , GTP-Binding Protein alpha Subunits/metabolism , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Domains , Receptors, Dopamine D1/chemistry , src-Family Kinases/metabolism
2.
Nat Chem Biol ; 16(12): 1440, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32807971

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Nat Chem Biol ; 16(12): 1343-1350, 2020 12.
Article in English | MEDLINE | ID: mdl-32778842

ABSTRACT

The adhesion G-protein-coupled receptor (GPCR) latrophilin 3 (ADGRL3) has been associated with increased risk of attention deficit hyperactivity disorder (ADHD) and substance use in human genetic studies. Knockdown in multiple species leads to hyperlocomotion and altered dopamine signaling. Thus, ADGRL3 is a potential target for treatment of neuropsychiatric disorders that involve dopamine dysfunction, but its basic signaling properties are poorly understood. Identification of adhesion GPCR signaling partners has been limited by a lack of tools to acutely activate these receptors in living cells. Here, we design a novel acute activation strategy to characterize ADGRL3 signaling by engineering a receptor construct in which we could trigger acute activation enzymatically. Using this assay, we found that ADGRL3 signals through G12/G13 and Gq, with G12/13 the most robustly activated. Gα12/13 is a new player in ADGRL3 biology, opening up unexplored roles for ADGRL3 in the brain. Our methodological advancements should be broadly useful in adhesion GPCR research.


Subject(s)
Activating Transcription Factor 6/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Peptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Activating Transcription Factor 6/agonists , Activating Transcription Factor 6/chemistry , Activating Transcription Factor 6/genetics , Animals , Arrestin/chemistry , Arrestin/genetics , Arrestin/metabolism , CRISPR-Cas Systems , Cell Engineering , GTP-Binding Protein alpha Subunits, G12-G13/chemistry , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression , HEK293 Cells , Humans , Kinetics , Mice , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
4.
Proc Natl Acad Sci U S A ; 116(3): 810-815, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30591558

ABSTRACT

Scaffold proteins tether and orient components of a signaling cascade to facilitate signaling. Although much is known about how scaffolds colocalize signaling proteins, it is unclear whether scaffolds promote signal amplification. Here, we used arrestin-3, a scaffold of the ASK1-MKK4/7-JNK3 cascade, as a model to understand signal amplification by a scaffold protein. We found that arrestin-3 exhibited >15-fold higher affinity for inactive JNK3 than for active JNK3, and this change involved a shift in the binding site following JNK3 activation. We used systems biochemistry modeling and Bayesian inference to evaluate how the activation of upstream kinases contributed to JNK3 phosphorylation. Our combined experimental and computational approach suggested that the catalytic phosphorylation rate of JNK3 at Thr-221 by MKK7 is two orders of magnitude faster than the corresponding phosphorylation of Tyr-223 by MKK4 with or without arrestin-3. Finally, we showed that the release of activated JNK3 was critical for signal amplification. Collectively, our data suggest a "conveyor belt" mechanism for signal amplification by scaffold proteins. This mechanism informs on a long-standing mystery for how few upstream kinase molecules activate numerous downstream kinases to amplify signaling.


Subject(s)
MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 10/metabolism , beta-Arrestin 2/metabolism , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase 7/metabolism , Models, Biological , Phosphorylation , Software
5.
Biochemistry ; 57(50): 6827-6837, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30525509

ABSTRACT

Members of the orthosomycin family of natural products are decorated polysaccharides with potent antibiotic activity and complex biosynthetic pathways. The defining feature of the orthosomycins is an orthoester linkage between carbohydrate moieties that is necessary for antibiotic activity and is likely formed by a family of conserved oxygenases. Everninomicins are octasaccharide orthosomycins produced by Micromonospora carbonacea that have two orthoester linkages and a methylenedioxy bridge, three features whose formation logically requires oxidative chemistry. Correspondingly, the evd gene cluster encoding everninomicin D encodes two monofunctional nonheme iron, α-ketoglutarate-dependent oxygenases and one bifunctional enzyme with an N-terminal methyltransferase domain and a C-terminal oxygenase domain. To investigate whether the activities of these domains are linked in the bifunctional enzyme EvdMO1, we determined the structure of the N-terminal methyltransferase domain to 1.1 Å and that of the full-length protein to 3.35 Å resolution. Both domains of EvdMO1 adopt the canonical folds of their respective superfamilies and are connected by a short linker. Each domain's active site is oriented such that it faces away from the other domain, and there is no evidence of a channel connecting the two. Our results support EvdMO1 working as a bifunctional enzyme with independent catalytic activities.


Subject(s)
Aminoglycosides/biosynthesis , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Methyltransferases/chemistry , Methyltransferases/metabolism , Micromonospora/enzymology , Oxygenases/chemistry , Oxygenases/metabolism , Amino Acid Sequence , Aminoglycosides/chemistry , Bacterial Proteins/genetics , Biosynthetic Pathways , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Gene Fusion , Genes, Bacterial , Methyltransferases/genetics , Micromonospora/genetics , Models, Molecular , Oxygenases/genetics , Protein Interaction Domains and Motifs , Sequence Homology, Amino Acid
6.
IUBMB Life ; 65(6): 479-86, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23512348

ABSTRACT

Discovered about a decade ago, obscurin (~720 kDa) is a member of a family of giant proteins expressed in striated muscle that are essential for normal muscle function. Much of what we understand about obscurin stems from its functions in cardiac and skeletal muscle. However, recent evidence has indicated that variants of obscurin ("obscurins") are expressed in diverse cell types, where they contribute to distinct cellular processes. Dysfunction or abrogation of obscurins has also been implicated in the development of several pathological conditions, including cardiac hypertrophy and cancer. Herein, we present an overview of obscurins with an emphasis on novel findings that demonstrate their heretofore-unsuspected importance in cell signaling and disease progression.


Subject(s)
Rho Guanine Nucleotide Exchange Factors/physiology , Animals , Cardiomyopathy, Hypertrophic, Familial/genetics , Humans , Muscle Development , Muscle, Skeletal/metabolism , Mutation, Missense , Neoplasms/genetics , Organ Specificity , Protein Interaction Maps , Protein Serine-Threonine Kinases
7.
FASEB J ; 26(7): 2764-75, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22441987

ABSTRACT

Obscurins (∼70 - 870 kDa), encoded by the single OBSCN gene, are cytoskeletal proteins originally identified in striated muscles with structural and regulatory roles. Recently, analysis of 13,023 genes in breast and colorectal cancers identified OBSCN as one of the most frequently mutated genes, implicating it in cancer formation. Herein we studied the expression profile of obscurins in breast, colon, and skin cancer cell lines and their involvement in cell survival. Immunoblot analysis demonstrated significant reduction of obscurin proteins [corrected] in cancer cells, resulting from decreased mRNA levels and/or the presence of mutant transcripts. In normal epithelium, obscurins localize in cytoplasmic puncta, the cell membrane, and the nucleus. Accordingly, subcellular fractionation demonstrated the presence of 2 novel nuclear isoforms of ∼110 and ∼120 kDa. Nontumorigenic MCF10A breast epithelial cells stably transduced with shRNAs targeting giant obscurins exhibited increased viability (∼30%) and reduced apoptosis (∼20%) following exposure to the DNA-damaging agent etoposide. Quantitative RT-PCR further indicated that the antiapoptotic genes BAG4 and HAX1 were up-regulated (1.5- and 1.4-fold, respectively), whereas initiator caspase-9 and death caspase-3 transcripts were down-regulated (0.8- and 0.6-fold, respectively). Our findings are the first to pinpoint critical roles for obscurins in cancer development by contributing to the regulation of cell survival.


Subject(s)
Breast/cytology , Breast/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/physiology , Muscle Proteins/genetics , Muscle Proteins/physiology , Apoptosis/genetics , Apoptosis/physiology , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/genetics , Cell Survival/physiology , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , DNA Damage , Epithelial Cells/cytology , Epithelial Cells/metabolism , Etoposide/toxicity , Female , Gene Expression , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/deficiency , Humans , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Rho Guanine Nucleotide Exchange Factors , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
8.
Methods Mol Biol ; 1957: 107-120, 2019.
Article in English | MEDLINE | ID: mdl-30919350

ABSTRACT

Nonvisual arrestins (arrestin-2/arrestin-3) interact with hundreds of G protein-coupled receptor (GPCR) subtypes and dozens of non-receptor signaling proteins. Here we describe the methods used to identify the interaction sites of arrestin-binding partners on arrestin-3 and the use of monofunctional individual arrestin-3 elements in cells. Our in vitro pull-down assay with purified proteins demonstrates that relatively few elements in arrestin engage each partner, whereas cell-based functional assays indicate that certain arrestin elements devoid of other functionalities can perform individual functions in living cells.


Subject(s)
Arrestin/metabolism , Biological Assay/methods , Protein Interaction Mapping/methods , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Immobilized Proteins/metabolism , Mice , Protein Binding , Recombinant Fusion Proteins/metabolism
9.
Cell Signal ; 63: 109366, 2019 11.
Article in English | MEDLINE | ID: mdl-31352007

ABSTRACT

Maternal embryonic leucine-zipper kinase (MELK) overexpression impacts survival and proliferation of multiple cancer types, most notably glioblastomas and breast cancer. This makes MELK an attractive molecular target for cancer therapy. Yet the molecular mechanisms underlying the involvement of MELK in tumorigenic processes are unknown. MELK participates in numerous protein-protein interactions that affect cell cycle, proliferation, apoptosis, and embryonic development. Here we used both in vitro and in-cell assays to identify a direct interaction between MELK and arrestin-3. A part of this interaction involves the MELK kinase domain, and we further show that the interaction between the MELK kinase domain and arrestin-3 decreases the number of cells in S-phase, as compared to cells expressing the MELK kinase domain alone. Thus, we describe a new mechanism of regulation of MELK function, which may contribute to the control of cell fate.


Subject(s)
Arrestins/chemistry , Arrestins/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , HEK293 Cells , Humans , Protein Binding , S Phase
10.
ACS Chem Biol ; 14(6): 1183-1194, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31058487

ABSTRACT

Extracellular signal-regulated kinases (ERK1/2) are mitogen-activated protein kinases (MAPKs) that play a pro-tumorigenic role in numerous cancers. ERK1/2 possess two protein-docking sites that are distinct from the active site: the D-recruitment site (DRS) and the F-recruitment site. These docking sites facilitate substrate recognition, intracellular localization, signaling specificity, and protein complex assembly. Targeting these sites on ERK in a therapeutic context may overcome many problems associated with traditional ATP-competitive inhibitors. Here, we identified a new class of inhibitors that target the ERK DRS by screening a synthetic combinatorial library of more than 30 million compounds. The screen detects the competitive displacement of a fluorescent peptide from the DRS of ERK2. The top molecular scaffold from the screen was optimized for structure-activity relationship by positional scanning of different functional groups. This resulted in 10 compounds with similar binding affinities and a shared core structure consisting of a tertiary amine hub with three functionalized cyclic guanidino branches. Compound 2507-1 inhibited ERK2 from phosphorylating a DRS-targeting substrate and prevented the phosphorylation of ERK2 by a constitutively active MEK1 (MAPK/ERK kinase 1) mutant. Interaction between an analogue, 2507-8, and the ERK2 DRS was confirmed by nuclear magnetic resonance and X-ray crystallography. 2507-8 forms critical interactions at the common docking domain residue Asp319 via an arginine-like moiety that is shared by all 10 hits, suggesting a common binding mode. The structural and biochemical insights reported here provide the basis for developing new ERK inhibitors that are not ATP-competitive but instead function by disrupting critical protein-protein interactions.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Binding Sites , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Activation , Guanidine/pharmacology , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/chemistry , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Substrate Specificity
11.
PLoS One ; 13(1): e0190842, 2018.
Article in English | MEDLINE | ID: mdl-29298356

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0088162.].

12.
Cell Signal ; 36: 98-107, 2017 08.
Article in English | MEDLINE | ID: mdl-28461104

ABSTRACT

Non-visual arrestins interact with hundreds of different G protein-coupled receptors (GPCRs). Here we show that by introducing mutations into elements that directly bind receptors, the specificity of arrestin-3 can be altered. Several mutations in the two parts of the central "crest" of the arrestin molecule, middle-loop and C-loop, enhanced or reduced arrestin-3 interactions with several GPCRs in receptor subtype and functional state-specific manner. For example, the Lys139Ile substitution in the middle-loop dramatically enhanced the binding to inactive M2 muscarinic receptor, so that agonist activation of the M2 did not further increase arrestin-3 binding. Thus, the Lys139Ile mutation made arrestin-3 essentially an activation-independent binding partner of M2, whereas its interactions with other receptors, including the ß2-adrenergic receptor and the D1 and D2 dopamine receptors, retained normal activation dependence. In contrast, the Ala248Val mutation enhanced agonist-induced arrestin-3 binding to the ß2-adrenergic and D2 dopamine receptors, while reducing its interaction with the D1 dopamine receptor. These mutations represent the first example of altering arrestin specificity via enhancement of the arrestin-receptor interactions rather than selective reduction of the binding to certain subtypes.


Subject(s)
Arrestins/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Arrestins/chemistry , COS Cells , Cattle , Chlorocebus aethiops , Conserved Sequence , HEK293 Cells , Humans , Lysine/metabolism , Mutant Proteins/metabolism , Mutation/genetics , Protein Binding , Protein Structure, Secondary , Rhodopsin/metabolism
13.
J Clin Invest ; 127(8): 2941-2945, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28650340

ABSTRACT

An increase in hepatic glucose production (HGP) represents a key feature of type 2 diabetes. This deficiency in metabolic control of glucose production critically depends on enhanced signaling through hepatic glucagon receptors (GCGRs). Here, we have demonstrated that selective inactivation of the GPCR-associated protein ß-arrestin 2 in hepatocytes of adult mice results in greatly increased hepatic GCGR signaling, leading to striking deficits in glucose homeostasis. However, hepatocyte-specific ß-arrestin 2 deficiency did not affect hepatic insulin sensitivity or ß-adrenergic signaling. Adult mice lacking ß-arrestin 1 selectively in hepatocytes did not show any changes in glucose homeostasis. Importantly, hepatocyte-specific overexpression of ß-arrestin 2 greatly reduced hepatic GCGR signaling and protected mice against the metabolic deficits caused by the consumption of a high-fat diet. Our data support the concept that strategies aimed at enhancing hepatic ß-arrestin 2 activity could prove useful for suppressing HGP for therapeutic purposes.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Liver/metabolism , Receptors, Glucagon/metabolism , beta-Arrestin 2/metabolism , Animals , COS Cells , Chlorocebus aethiops , Diet, High-Fat , Gene Deletion , Glucagon/metabolism , Hepatocytes/metabolism , Homeostasis/physiology , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , beta-Arrestin 1/genetics
14.
Nat Commun ; 8(1): 1427, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127291

ABSTRACT

A unique aspect of arrestin-3 is its ability to support both receptor-dependent and receptor-independent signaling. Here, we show that inositol hexakisphosphate (IP6) is a non-receptor activator of arrestin-3 and report the structure of IP6-activated arrestin-3 at 2.4-Å resolution. IP6-activated arrestin-3 exhibits an inter-domain twist and a displaced C-tail, hallmarks of active arrestin. IP6 binds to the arrestin phosphate sensor, and is stabilized by trimerization. Analysis of the trimerization surface, which is also the receptor-binding surface, suggests a feature called the finger loop as a key region of the activation sensor. We show that finger loop helicity and flexibility may underlie coupling to hundreds of diverse receptors and also promote arrestin-3 activation by IP6. Importantly, we show that effector-binding sites on arrestins have distinct conformations in the basal and activated states, acting as switch regions. These switch regions may work with the inter-domain twist to initiate and direct arrestin-mediated signaling.


Subject(s)
Arrestins/chemistry , Arrestins/metabolism , Amino Acid Sequence , Animals , Arrestins/genetics , Binding Sites , Cattle , Crystallography, X-Ray , Humans , Mitogen-Activated Protein Kinase 10/metabolism , Models, Molecular , Phytic Acid/metabolism , Protein Conformation , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
15.
Nat Commun ; 8: 14295, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28145434

ABSTRACT

ß-arrestins are critical signalling molecules that regulate many fundamental physiological functions including the maintenance of euglycemia and peripheral insulin sensitivity. Here we show that inactivation of the ß-arrestin-2 gene, barr2, in ß-cells of adult mice greatly impairs insulin release and glucose tolerance in mice fed with a calorie-rich diet. Both glucose and KCl-induced insulin secretion and calcium responses were profoundly reduced in ß-arrestin-2 (barr2) deficient ß-cells. In human ß-cells, barr2 knockdown abolished glucose-induced insulin secretion. We also show that the presence of barr2 is essential for proper CAMKII function in ß-cells. Importantly, overexpression of barr2 in ß-cells greatly ameliorates the metabolic deficits displayed by mice consuming a high-fat diet. Thus, our data identify barr2 as an important regulator of ß-cell function, which may serve as a new target to improve ß-cell function.


Subject(s)
Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Signal Transduction/genetics , beta-Arrestin 2/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Cell Line, Tumor , Diet, High-Fat , Gene Expression , Humans , Insulin/metabolism , Insulin Secretion , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , beta-Arrestin 2/metabolism
16.
Sci Rep ; 6: 21025, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26868142

ABSTRACT

Three-kinase mitogen-activated protein kinase (MAPK) signaling cascades are present in virtually all eukaryotic cells. MAPK cascades are organized by scaffold proteins, which assemble cognate kinases into productive signaling complexes. Arrestin-3 facilitates JNK activation in cells, and a short 25-residue arrestin-3 peptide was identified as the critical JNK3-binding element. Here we demonstrate that this peptide also binds MKK4, MKK7, and ASK1, which are upstream JNK3-activating kinases. This peptide is sufficient to enhance JNK3 activity in cells. A homologous arrestin-2 peptide, which differs only in four positions, binds MKK4, but not MKK7 or JNK3, and is ineffective in cells at enhancing activation of JNK3. The arrestin-3 peptide is the smallest MAPK scaffold known. This peptide or its mimics can regulate MAPKs, affecting cellular decisions to live or die.


Subject(s)
Enzyme Activators , Mitogen-Activated Protein Kinase 10/metabolism , Peptides , beta-Arrestin 1/chemistry , beta-Arrestin 2/chemistry , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation/drug effects , Enzyme Activators/chemical synthesis , Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Humans , Mitogen-Activated Protein Kinase 10/genetics , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology
17.
PLoS One ; 9(2): e88162, 2014.
Article in English | MEDLINE | ID: mdl-24516603

ABSTRACT

Obscurins comprise a family of proteins originally identified in striated muscles, where they play essential roles in myofibrillogenesis, cytoskeletal organization, and Ca(2+) homeostasis. They are encoded by the single OBSCN gene, and are composed of tandem adhesion domains and signaling motifs. To date, two giant obscurin isoforms have been described in detail that differ only at the extreme COOH-terminus; while obscurin-A (∼720 kDa) contains a non-modular COOH-terminus that harbors binding sites for the adaptor proteins ankyrins, obscurin-B (∼870 kDa) contains two COOH-terminal serine-threonine kinase domains preceded by adhesion motifs. Besides the two known giant obscurins, a thorough search of transcript databases suggests that complex alternative splicing of the obscurin transcript results in the generation of additional giant as well as small isoforms with molecular masses ranging between ∼50-970 kDa. These novel isoforms share common domains with the characterized isoforms, but also contain unique regions. Using a panel of highly specific antibodies directed against epitopes spanning the entire length of giant obscurins, we employed western blotting and immunohistochemistry to perform a systematic and comprehensive characterization of the expression profile of obscurins in muscle and non-muscle tissues. Our studies demonstrate for the first time that obscurins are not restricted to striated muscles, but are abundantly expressed in several tissues and organs including brain, skin, kidney, liver, spleen, and lung. While some obscurin isoforms are ubiquitously expressed, others are preferentially present in specific tissues and organs. Moreover, obscurins are present in select structures and cell types where they assume nuclear, cytosolic, and membrane distributions. Given the ubiquitous expression of some obscurins, along with the preferential expression of others, it becomes apparent that obscurins may play common and unique roles, respectively, in the regulation and maintenance of cell homeostasis in various tissues and organs throughout the body.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Muscle Proteins/metabolism , Muscles/metabolism , Animals , Epitopes/immunology , Female , Gene Expression Profiling , Humans , Mice, Inbred C57BL , Muscle, Striated/metabolism , Organ Specificity/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases , Protein Transport , Rats, Sprague-Dawley , Rho Guanine Nucleotide Exchange Factors , Subcellular Fractions/metabolism
18.
Oncotarget ; 5(18): 8558-68, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25261370

ABSTRACT

Obscurins are RhoGEF-containing proteins whose downregulation has been implicated in the development and progression of breast cancer. Herein, we aim to elucidate the mechanism for increased motility of obscurin-deficient cells. We show that shRNA-mediated obscurin downregulation in MCF10A cells leads to >50% reduction in RhoA activity relative to scramble control (shCtrl), as well as decreased phosphorylation of RhoA effectors, including myosin light chain phosphatase, myosin light chain, lim kinase, and cofilin, in both attached and suspended cells. These alterations result in decreased actomyosin contractility, allowing suspended cells to escape detachment-induced apoptosis. Moreover, ~40% of shObsc-expressing cells, but only ~10% of shCtrl-expressing cells, extend microtentacles, tubulin-based projections that mediate the attachment of circulating tumor cells to endothelium. Indeed, we show that MCF10A cells expressing shObsc attach in vitro more readily than shCtrl cells, an advantage that persists following taxane exposure. Overall, our data suggest that loss of obscurins may represent a substantial selective advantage for breast epithelial cells during metastasis, and that treatment with paclitaxel may exacerbate this advantage by preferentially allowing obscurin-deficient, stem-like cells to attach to the endothelium of distant sites, a first step towards colonizing metastatic tumors.


Subject(s)
Breast Neoplasms/metabolism , Cell Adhesion , Cell Surface Extensions/metabolism , Epithelial Cells/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement , Cell Surface Extensions/pathology , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Paclitaxel/pharmacology , Protein Serine-Threonine Kinases , RNA Interference , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction/drug effects , Time Factors , Transfection , Tubulin/metabolism , Tubulin Modulators/pharmacology , rhoA GTP-Binding Protein/genetics
19.
PLoS One ; 5(9)2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20844765

ABSTRACT

BACKGROUND: Several short antimicrobial peptides that are rich in tryptophan and arginine residues were designed with a series of simple modifications such as end capping and cyclization. The two sets of hexapeptides are based on the Trp- and Arg-rich primary sequences from the "antimicrobial centre" of bovine lactoferricin as well as an antimicrobial sequence obtained through the screening of a hexapeptide combinatorial library. METHODOLOGY/PRINCIPAL FINDINGS: HPLC, mass spectrometry and antimicrobial assays were carried out to explore the consequences of the modifications on the serum stability and microbicidal activity of the peptides. The results show that C-terminal amidation increases the antimicrobial activity but that it makes little difference to its proteolytic degradation in human serum. On the other hand, N-terminal acetylation decreases the peptide activities but significantly increases their protease resistance. Peptide cyclization of the hexameric peptides was found to be highly effective for both serum stability and antimicrobial activity. However the two cyclization strategies employed have different effects, with disulfide cyclization resulting in more active peptides while backbone cyclization results in more proteolytically stable peptides. However, the benefit of backbone cyclization did not extend to longer 11-mer peptides derived from the same region of lactoferricin. Mass spectrometry data support the serum stability assay results and allowed us to determine preferred proteolysis sites in the peptides. Furthermore, isothermal titration calorimetry experiments showed that the peptides all had weak interactions with albumin, the most abundant protein in human serum. CONCLUSIONS/SIGNIFICANCE: Taken together, the results provide insight into the behavior of the peptides in human serum and will therefore aid in advancing antimicrobial peptide design towards systemic applications.


Subject(s)
Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Arginine/chemistry , Tryptophan/chemistry , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/blood , Antimicrobial Cationic Peptides/pharmacology , Bacteria/drug effects , Humans , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL