Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
Curr Opin Chem Biol ; 74: 102279, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36966700

RESUMO

Eukaryotes depend upon the proper localization, accumulation, and release of intracellular Ca2+. This is regulated through specialized cellular compartments, signaling pathways, and Ca2+-binding proteins and channels. Cytosolic and extracellular signaling governing intracellular Ca2+ stores are well explored. However, regulatory signals within Ca2+ storage organelles like the endoplasmic/sarcoplasmic reticulum are not well understood. This is due to a lack of identified signaling molecules - like protein kinases - within these compartments, limited information on their regulation, and incomplete understanding of mechanisms involving modified substrates. Here we review recent advances in intralumenal signaling focusing on the secretory pathway protein kinase FAM20C and its regulation, Ca2+-binding protein substrates, and potential mechanisms through which FAM20C may regulate Ca2+ storage.


Assuntos
Cálcio , Retículo Sarcoplasmático , Retículo Sarcoplasmático/metabolismo , Cálcio/metabolismo , Via Secretória , Retículo Endoplasmático/metabolismo , Fosfotransferases/metabolismo
2.
Nat Chem Biol ; 18(10): 1076-1086, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35788180

RESUMO

The Hippo pathway plays a key role in development, organ size control and tissue homeostasis, and its dysregulation contributes to cancer. The LATS tumor suppressor kinases phosphorylate and inhibit the YAP/TAZ transcriptional co-activators to suppress gene expression and cell growth. Through a screen of marine natural products, we identified microcolin B (MCB) as a Hippo activator that preferentially kills YAP-dependent cancer cells. Structure-activity optimization yielded more potent MCB analogs, which led to the identification of phosphatidylinositol transfer proteins α and ß (PITPα/ß) as the direct molecular targets. We established a critical role of PITPα/ß in regulating LATS and YAP. Moreover, we showed that PITPα/ß influence the Hippo pathway via plasma membrane phosphatidylinositol-4-phosphate. This study uncovers a previously unrecognized role of PITPα/ß in Hippo pathway regulation and as potential cancer therapeutic targets.


Assuntos
Produtos Biológicos , Neoplasias , Humanos , Via de Sinalização Hippo , Fosfatidilinositóis , Proteínas de Transferência de Fosfolipídeos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Fatores de Transcrição/metabolismo
3.
J Biol Chem ; 296: 100267, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33759783

RESUMO

The study of extracellular phosphorylation was initiated in late 19th century when the secreted milk protein, casein, and egg-yolk protein, phosvitin, were shown to be phosphorylated. However, it took more than a century to identify Fam20C, which phosphorylates both casein and phosvitin under physiological conditions. This kinase, along with its family members Fam20A and Fam20B, defined a new family with altered amino acid sequences highly atypical from the canonical 540 kinases comprising the kinome. Fam20B is a glycan kinase that phosphorylates xylose residues and triggers peptidoglycan biosynthesis, a role conserved from sponges to human. The protein kinase, Fam20C, conserved from nematodes to humans, phosphorylates well over 100 substrates in the secretory pathway with overall functions postulated to encompass endoplasmic reticulum homeostasis, nutrition, cardiac function, coagulation, and biomineralization. The preferred phosphorylation motif of Fam20C is SxE/pS, and structural studies revealed that related member Fam20A allosterically activates Fam20C by forming a heterodimeric/tetrameric complex. Fam20A, a pseudokinase, is observed only in vertebrates. Loss-of-function genetic alterations in the Fam20 family lead to human diseases such as amelogenesis imperfecta, nephrocalcinosis, lethal and nonlethal forms of Raine syndrome with major skeletal defects, and altered phosphate homeostasis. Together, these three members of the Fam20 family modulate a diverse network of secretory pathway components playing crucial roles in health and disease. The overarching theme of this review is to highlight the progress that has been made in the emerging field of extracellular phosphorylation and the key roles secretory pathway kinases play in an ever-expanding number of cellular processes.


Assuntos
Caseína Quinase I/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Caseína Quinase I/química , Retículo Endoplasmático/metabolismo , Proteínas da Matriz Extracelular/química , Homeostase , Humanos , Miocárdio/metabolismo , Fosforilação , Via Secretória , Transdução de Sinais , Relação Estrutura-Atividade , Especificidade por Substrato
4.
Cell Death Differ ; 28(5): 1563-1578, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33268814

RESUMO

To survive proteotoxic stress, cancer cells activate the proteotoxic-stress response pathway, which is controlled by the transcription factor heat shock factor 1 (HSF1). This pathway supports cancer initiation, cancer progression and chemoresistance and thus is an attractive therapeutic target. As developing inhibitors against transcriptional regulators, such as HSF1 is challenging, the identification and targeting of upstream regulators of HSF1 present a tractable alternative strategy. Here we demonstrate that in triple-negative breast cancer (TNBC) cells, the dual specificity tyrosine-regulated kinase 2 (DYRK2) phosphorylates HSF1, promoting its nuclear stability and transcriptional activity. DYRK2 depletion reduces HSF1 activity and sensitises TNBC cells to proteotoxic stress. Importantly, in tumours from TNBC patients, DYRK2 levels positively correlate with active HSF1 and associates with poor prognosis, suggesting that DYRK2 could be promoting TNBC. These findings identify DYRK2 as a key modulator of the HSF1 transcriptional programme and a potential therapeutic target.


Assuntos
Fatores de Transcrição de Choque Térmico/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Humanos , Prognóstico , Fatores de Transcrição/metabolismo , Transfecção , Quinases Dyrk
5.
Elife ; 92020 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-32975514

RESUMO

Matriglycan [-GlcA-ß1,3-Xyl-α1,3-]n serves as a scaffold in many tissues for extracellular matrix proteins containing laminin-G domains including laminin, agrin, and perlecan. Like-acetyl-glucosaminyltransferase 1 (LARGE1) synthesizes and extends matriglycan on α-dystroglycan (α-DG) during skeletal muscle differentiation and regeneration; however, the mechanisms which regulate matriglycan elongation are unknown. Here, we show that Protein O-Mannose Kinase (POMK), which phosphorylates mannose of core M3 (GalNAc-ß1,3-GlcNAc-ß1,4-Man) preceding matriglycan synthesis, is required for LARGE1-mediated generation of full-length matriglycan on α-DG (~150 kDa). In the absence of Pomk gene expression in mouse skeletal muscle, LARGE1 synthesizes a very short matriglycan resulting in a ~ 90 kDa α-DG which binds laminin but cannot prevent eccentric contraction-induced force loss or muscle pathology. Solution NMR spectroscopy studies demonstrate that LARGE1 directly interacts with core M3 and binds preferentially to the phosphorylated form. Collectively, our study demonstrates that phosphorylation of core M3 by POMK enables LARGE1 to elongate matriglycan on α-DG, thereby preventing muscular dystrophy.


Assuntos
Distroglicanas/metabolismo , Expressão Gênica , Músculo Esquelético/fisiologia , N-Acetilglucosaminiltransferases/genética , Proteínas Quinases/genética , Animais , Masculino , Manose/química , Camundongos , N-Acetilglucosaminiltransferases/metabolismo , Fosforilação , Proteínas Quinases/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(1): 328-336, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31843888

RESUMO

The fundamental importance of the 26S proteasome in health and disease suggests that its function must be finely controlled, and yet our knowledge about proteasome regulation remains limited. Posttranslational modifications, especially phosphorylation, of proteasome subunits have been shown to impact proteasome function through different mechanisms, although the vast majority of proteasome phosphorylation events have not been studied. Here, we have characterized 1 of the most frequently detected proteasome phosphosites, namely Ser361 of Rpn1, a base subunit of the 19S regulatory particle. Using a variety of approaches including CRISPR/Cas9-mediated gene editing and quantitative mass spectrometry, we found that loss of Rpn1-S361 phosphorylation reduces proteasome activity, impairs cell proliferation, and causes oxidative stress as well as mitochondrial dysfunction. A screen of the human kinome identified several kinases including PIM1/2/3 that catalyze S361 phosphorylation, while its level is reversibly controlled by the proteasome-resident phosphatase, UBLCP1. Mechanistically, Rpn1-S361 phosphorylation is required for proper assembly of the 26S proteasome, and we have utilized a genetic code expansion system to directly demonstrate that S361-phosphorylated Rpn1 more readily forms a precursor complex with Rpt2, 1 of the first steps of 19S base assembly. These findings have revealed a prevalent and biologically important mechanism governing proteasome formation and function.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Subunidades Proteicas/metabolismo , Animais , Sistemas CRISPR-Cas/genética , Linhagem Celular , Ensaios Enzimáticos , Técnicas de Introdução de Genes , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/metabolismo , Proteínas Nucleares/genética , Estresse Oxidativo , Fosfoproteínas Fosfatases/genética , Fosforilação/fisiologia , Complexo de Endopeptidases do Proteassoma/genética , Proteínas Serina-Treonina Quinases/genética , Subunidades Proteicas/genética , RNA Interferente Pequeno/metabolismo , Serina/metabolismo , Transativadores/genética , Transativadores/metabolismo
7.
Proc Natl Acad Sci U S A ; 116(49): 24881-24891, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31754034

RESUMO

Dependence on the 26S proteasome is an Achilles' heel for triple-negative breast cancer (TNBC) and multiple myeloma (MM). The therapeutic proteasome inhibitor, bortezomib, successfully targets MM but often leads to drug-resistant disease relapse and fails in breast cancer. Here we show that a 26S proteasome-regulating kinase, DYRK2, is a therapeutic target for both MM and TNBC. Genome editing or small-molecule mediated inhibition of DYRK2 significantly reduces 26S proteasome activity, bypasses bortezomib resistance, and dramatically delays in vivo tumor growth in MM and TNBC thereby promoting survival. We further characterized the ability of LDN192960, a potent and selective DYRK2-inhibitor, to alleviate tumor burden in vivo. The drug docks into the active site of DYRK2 and partially inhibits all 3 core peptidase activities of the proteasome. Our results suggest that targeting 26S proteasome regulators will pave the way for therapeutic strategies in MM and TNBC.


Assuntos
Bortezomib/farmacologia , Processos Neoplásicos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , TYK2 Quinase/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , ATPases Associadas a Diversas Atividades Celulares/genética , Animais , Linhagem Celular Tumoral , Feminino , Edição de Genes , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mieloma Múltiplo , Fosforilação , Complexo de Endopeptidases do Proteassoma/genética , Inibidores de Proteassoma/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Neoplasias de Mama Triplo Negativas/patologia , Quinases Dyrk
8.
J Cell Biol ; 218(11): 3795-3811, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31541016

RESUMO

Fam20C is a secreted protein kinase mutated in Raine syndrome, a human skeletal disorder. In vertebrates, bone and enamel proteins are major Fam20C substrates. However, Fam20 kinases are conserved in invertebrates lacking bone and enamel, suggesting other ancestral functions. We show that FAMK-1, the Caenorhabditis elegans Fam20C orthologue, contributes to fertility, embryogenesis, and development. These functions are not fulfilled when FAMK-1 is retained in the early secretory pathway. During embryogenesis, FAMK-1 maintains intercellular partitions and prevents multinucleation; notably, temperature elevation or lowering cortical stiffness reduces requirement for FAMK-1 in these contexts. FAMK-1 is expressed in multiple adult tissues that undergo repeated mechanical strain, and selective expression in the spermatheca restores fertility. Informatic, biochemical, and functional analysis implicate lectins as FAMK-1 substrates. These findings suggest that FAMK-1 phosphorylation of substrates, including lectins, in the late secretory pathway is important in embryonic and tissue contexts where cells are subjected to mechanical strain.


Assuntos
Caenorhabditis elegans/enzimologia , Caseína Quinase I/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Caseína Quinase I/genética , Células HEK293 , Humanos
9.
J Biol Chem ; 294(5): 1638-1642, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30710011

RESUMO

Herb Tabor was the Editor-in-Chief of the Journal of Biological Chemistry (JBC) spanning the years 1971-2010. This year, Herb turns 100. What do you give a person turning 100? Our answer to this question was to dedicate two of our favorite JBC papers to Herb. Both of these papers focus on reversible phosphorylation, which we briefly review. In addition, we delve into a new finding that centers around a novel family of secreted kinases, suggesting that there are many new and exciting discoveries yet to explore.


Assuntos
Bioquímica/história , Publicações Periódicas como Assunto , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , História do Século XX , História do Século XXI , Humanos , Fosforilação
10.
Elife ; 72018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30520731

RESUMO

Ca2+ signaling is important for many cellular and physiological processes, including cardiac function. Although sarcoplasmic reticulum (SR) proteins involved in Ca2+ signaling have been shown to be phosphorylated, the biochemical and physiological roles of protein phosphorylation within the lumen of the SR remain essentially uncharacterized. Our laboratory recently identified an atypical protein kinase, Fam20C, which is uniquely localized to the secretory pathway lumen. Here, we show that Fam20C phosphorylates several SR proteins involved in Ca2+ signaling, including calsequestrin2 and Stim1, whose biochemical activities are dramatically regulated by Fam20C mediated phosphorylation. Notably, phosphorylation of Stim1 by Fam20C enhances Stim1 activation and store-operated Ca2+ entry. Physiologically, mice with Fam20c ablated in cardiomyocytes develop heart failure following either aging or induced pressure overload. We extended these observations to show that non-muscle cells lacking Fam20C display altered ER Ca2+ signaling. Overall, we show that Fam20C plays an overarching role in ER/SR Ca2+ homeostasis and cardiac pathophysiology.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Calsequestrina/genética , Proteínas da Matriz Extracelular/genética , Insuficiência Cardíaca/genética , Molécula 1 de Interação Estromal/genética , Animais , Cálcio/química , Cálcio/metabolismo , Sinalização do Cálcio/genética , Proteínas de Ligação ao Cálcio/química , Calsequestrina/química , Retículo Endoplasmático/química , Retículo Endoplasmático/genética , Proteínas da Matriz Extracelular/química , Insuficiência Cardíaca/patologia , Homeostase , Humanos , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação , Fosfotransferases/genética , Retículo Sarcoplasmático/química , Retículo Sarcoplasmático/genética , Via Secretória/genética , Molécula 1 de Interação Estromal/química
11.
Biophys J ; 115(12): 2327-2335, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30527445

RESUMO

Phosphoproteomics studies have reported phosphorylation at multiple sites within collagen, raising the possibility that these post-translational modifications regulate the physical or biological properties of collagen. In this study, molecular dynamics simulations and experimental studies were carried out on model peptides to establish foundational principles of phosphorylation of Ser residues in collagen. A (Gly-Xaa-Yaa)11 peptide was designed to include a Ser-containing sequence from type I collagen that was reported to be phosphorylated. The physiological kinase involved in collagen phosphorylation is not known. In vitro studies showed that a model kinase ERK1 (extracellular signal-regulated protein kinase 1) would phosphorylate Ser within the consensus sequence if the collagen-like peptide is in the denatured state but not in the triple-helical state. The peptide was not a substrate for FAM20C, a kinase present in the secretory pathway, which has been shown to phosphorylate many extracellular matrix proteins. The unfolded single chain (Gly-Xaa-Yaa)11 peptide containing phosphoSer was able to refold to form a stable triple helix but at a reduced folding rate and with a small decrease in thermal stability relative to the nonphosphorylated peptide at neutral pH. These biophysical studies on model peptides provide a basis for investigations into the physiological consequences of collagen phosphorylation and the application of phosphorylation to regulate the properties of collagen biomaterials.


Assuntos
Colágeno Tipo I/química , Colágeno Tipo I/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Serina/metabolismo , Sequência de Aminoácidos , Simulação de Dinâmica Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosforilação , Conformação Proteica em alfa-Hélice , Dobramento de Proteína , Estabilidade Proteica
12.
Proc Natl Acad Sci U S A ; 115(32): 8155-8160, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-29987021

RESUMO

Curcumin, the active ingredient in Curcuma longa, has been in medicinal use since ancient times. However, the therapeutic targets and signaling cascades modulated by curcumin have been enigmatic despite extensive research. Here we identify dual-specificity tyrosine-regulated kinase 2 (DYRK2), a positive regulator of the 26S proteasome, as a direct target of curcumin. Curcumin occupies the ATP-binding pocket of DYRK2 in the cocrystal structure, and it potently and specifically inhibits DYRK2 over 139 other kinases tested in vitro. As a result, curcumin diminishes DYRK2-mediated 26S proteasome phosphorylation in cells, leading to reduced proteasome activity and impaired cell proliferation. Interestingly, curcumin synergizes with the therapeutic proteasome inhibitor carfilzomib to induce apoptosis in a variety of proteasome-addicted cancer cells, while this drug combination exhibits modest to no cytotoxicity to noncancerous cells. In a breast cancer xenograft model, curcumin treatment significantly reduces tumor burden in immunocompromised mice, showing a similar antitumor effect as CRISPR/Cas9-mediated DYRK2 depletion. These results reveal an unexpected role of curcumin in DYRK2-proteasome inhibition and provide a proof-of-concept that pharmacological manipulation of proteasome regulators may offer new opportunities for anticancer treatment.


Assuntos
Antineoplásicos/farmacologia , Curcumina/farmacologia , Neoplasias/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Sistemas CRISPR-Cas , Proliferação de Células/efeitos dos fármacos , Cristalografia por Raios X , Curcumina/uso terapêutico , Sinergismo Farmacológico , Feminino , Edição de Genes/métodos , Técnicas de Inativação de Genes/métodos , Células HEK293 , Humanos , Concentração Inibidora 50 , Camundongos , Neoplasias/patologia , Oligopeptídeos/farmacologia , Inibidores de Proteassoma/farmacologia , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases Dyrk
13.
Nat Commun ; 9(1): 1218, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29572475

RESUMO

The Fam20 proteins are novel kinases that phosphorylate secreted proteins and proteoglycans. Fam20C phosphorylates hundreds of secreted proteins and is activated by the pseudokinase Fam20A. Fam20B phosphorylates a xylose residue to regulate proteoglycan synthesis. Despite these wide-ranging and important functions, the molecular and structural basis for the regulation and substrate specificity of these kinases are unknown. Here we report molecular characterizations of all three Fam20 kinases, and show that Fam20C is activated by the formation of an evolutionarily conserved homodimer or heterodimer with Fam20A. Fam20B has a unique active site for recognizing Galß1-4Xylß1, the initiator disaccharide within the tetrasaccharide linker region of proteoglycans. We further show that in animals the monomeric Fam20B preceded the appearance of the dimeric Fam20C, and the dimerization trait of Fam20C emerged concomitantly with a change in substrate specificity. Our results provide comprehensive structural, biochemical, and evolutionary insights into the function of the Fam20 kinases.


Assuntos
Caseína Quinase I/química , Proteínas da Matriz Extracelular/química , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Domínio Catalítico , Cristalografia por Raios X , Humanos , Ligação de Hidrogênio , Insetos , Mutação , Fosforilação , Filogenia , Polissacarídeos/química , Multimerização Proteica , Proteoglicanas/química , Especificidade por Substrato , Xilose/química
14.
Proc Natl Acad Sci U S A ; 114(34): 9098-9103, 2017 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-28784772

RESUMO

Precise Ca cycling through the sarcoplasmic reticulum (SR), a Ca storage organelle, is critical for proper cardiac muscle function. This cycling initially involves SR release of Ca via the ryanodine receptor, which is regulated by its interacting proteins junctin and triadin. The sarco/endoplasmic reticulum Ca ATPase (SERCA) pump then refills SR Ca stores. Histidine-rich Ca-binding protein (HRC) resides in the lumen of the SR, where it contributes to the regulation of Ca cycling by protecting stressed or failing hearts. The common Ser96Ala human genetic variant of HRC strongly correlates with life-threatening ventricular arrhythmias in patients with idiopathic dilated cardiomyopathy. However, the underlying molecular pathways of this disease remain undefined. Here, we demonstrate that family with sequence similarity 20C (Fam20C), a recently characterized protein kinase in the secretory pathway, phosphorylates HRC on Ser96. HRC Ser96 phosphorylation was confirmed in cells and human hearts. Furthermore, a Ser96Asp HRC variant, which mimics constitutive phosphorylation of Ser96, diminished delayed aftercontractions in HRC null cardiac myocytes. This HRC phosphomimetic variant was also able to rescue the aftercontractions elicited by the Ser96Ala variant, demonstrating that phosphorylation of Ser96 is critical for the cardioprotective function of HRC. Phosphorylation of HRC on Ser96 regulated the interactions of HRC with both triadin and SERCA2a, suggesting a unique mechanism for regulation of SR Ca homeostasis. This demonstration of the role of Fam20C-dependent phosphorylation in heart disease will open new avenues for potential therapeutic approaches against arrhythmias.


Assuntos
Arritmias Cardíacas/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Caseína Quinase I/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Sequência de Aminoácidos , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/prevenção & controle , Proteínas de Ligação ao Cálcio/genética , Caseína Quinase I/genética , Linhagem Celular Tumoral , Células Cultivadas , Proteínas da Matriz Extracelular/genética , Humanos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Miócitos Cardíacos/metabolismo , Fosforilação , Ratos , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Serina/genética , Serina/metabolismo
15.
Elife ; 62017 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-28432788

RESUMO

Mutations in FAM20A cause tooth enamel defects known as Amelogenesis Imperfecta (AI) and renal calcification. We previously showed that Fam20A is a secretory pathway pseudokinase and allosterically activates the physiological casein kinase Fam20C to phosphorylate secreted proteins important for biomineralization (Cui et al., 2015). Here we report the nucleotide-free and ATP-bound structures of Fam20A. Fam20A exhibits a distinct disulfide bond pattern mediated by a unique insertion region. Loss of this insertion due to abnormal mRNA splicing interferes with the structure and function of Fam20A, resulting in AI. Fam20A binds ATP in the absence of divalent cations, and strikingly, ATP is bound in an inverted orientation compared to other kinases. Fam20A forms a dimer in the crystal, and residues in the dimer interface are critical for Fam20C activation. Together, these results provide structural insights into the function of Fam20A and shed light on the mechanism by which Fam20A mutations cause disease.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas do Esmalte Dentário/química , Proteínas do Esmalte Dentário/metabolismo , Dissulfetos/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Multimerização Proteica
16.
Sci Signal ; 10(474)2017 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-28400531

RESUMO

Protein phosphatases are the essential opposite to protein kinases; together, these enzymes regulate all protein phosphorylation and most cellular processes. To better understand the global roles of protein phosphorylation, we cataloged the human protein phosphatome, composed of 189 known and predicted human protein phosphatase genes. We also identified 79 protein phosphatase pseudogenes or retrogenes, some of which may have residual function. We traced the origin and diversity of phosphatases by building protein phosphatomes for eight other eukaryotes, from the protist Dictyostelium to the sea urchin. We classified protein phosphatases from all nine species into a hierarchy of 10 protein folds, 21 families, and 178 subfamilies. We found that >80% of the 101 human subfamilies were conserved across the animal kingdom, but show substantial differences in evolution, including losses and expansions of individual subfamilies and changes in accessory domains. Protein phosphatases show similar evolutionary dynamics to those of kinases, with substantial losses in major model organisms. Sequence analysis predicts that 26 human protein phosphatase domains are catalytically disabled and that this disability is mostly conserved across orthologs. This genomic and evolutionary perspective on protein phosphatases provides a framework for global analysis of protein phosphorylation throughout the animal kingdom.


Assuntos
Evolução Molecular , Genômica/métodos , Fosfoproteínas Fosfatases/genética , Filogenia , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Animais , Domínio Catalítico/genética , Eucariotos/classificação , Eucariotos/enzimologia , Eucariotos/genética , Humanos , Fosfoproteínas Fosfatases/classificação , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteoma/genética , Proteoma/metabolismo , Proteômica/métodos , Pseudogenes/genética
17.
Mol Cell Proteomics ; 16(5): 840-854, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28292943

RESUMO

The 26S proteasome is the macromolecular machine responsible for ATP/ubiquitin dependent degradation. As aberration in proteasomal degradation has been implicated in many human diseases, structural analysis of the human 26S proteasome complex is essential to advance our understanding of its action and regulation mechanisms. In recent years, cross-linking mass spectrometry (XL-MS) has emerged as a powerful tool for elucidating structural topologies of large protein assemblies, with its unique capability of studying protein complexes in cells. To facilitate the identification of cross-linked peptides, we have previously developed a robust amine reactive sulfoxide-containing MS-cleavable cross-linker, disuccinimidyl sulfoxide (DSSO). To better understand the structure and regulation of the human 26S proteasome, we have established new DSSO-based in vivo and in vitro XL-MS workflows by coupling with HB-tag based affinity purification to comprehensively examine protein-protein interactions within the 26S proteasome. In total, we have identified 447 unique lysine-to-lysine linkages delineating 67 interprotein and 26 intraprotein interactions, representing the largest cross-link dataset for proteasome complexes. In combination with EM maps and computational modeling, the architecture of the 26S proteasome was determined to infer its structural dynamics. In particular, three proteasome subunits Rpn1, Rpn6, and Rpt6 displayed multiple conformations that have not been previously reported. Additionally, cross-links between proteasome subunits and 15 proteasome interacting proteins including 9 known and 6 novel ones have been determined to demonstrate their physical interactions at the amino acid level. Our results have provided new insights on the dynamics of the 26S human proteasome and the methodologies presented here can be applied to study other protein complexes.


Assuntos
Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Linhagem Celular , Humanos , Modelos Moleculares , Ligação Proteica , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem
18.
Elife ; 52016 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-27879205

RESUMO

The 'pseudokinase' SgK196 is a protein O-mannose kinase (POMK) that catalyzes an essential phosphorylation step during biosynthesis of the laminin-binding glycan on α-dystroglycan. However, the catalytic mechanism underlying this activity remains elusive. Here we present the crystal structure of Danio rerio POMK in complex with Mg2+ ions, ADP, aluminum fluoride, and the GalNAc-ß3-GlcNAc-ß4-Man trisaccharide substrate, thereby providing a snapshot of the catalytic transition state of this unusual kinase. The active site of POMK is established by residues located in non-canonical positions and is stabilized by a disulfide bridge. GalNAc-ß3-GlcNAc-ß4-Man is recognized by a surface groove, and the GalNAc-ß3-GlcNAc moiety mediates the majority of interactions with POMK. Expression of various POMK mutants in POMK knockout cells further validated the functional requirements of critical residues. Our results provide important insights into the ability of POMK to function specifically as a glycan kinase, and highlight the structural diversity of the human kinome.


Assuntos
Difosfato de Adenosina/química , Distroglicanas/química , Proteínas de Peixes/química , Magnésio/química , Manose/química , Proteínas Quinases/química , Trissacarídeos/química , Difosfato de Adenosina/metabolismo , Compostos de Alumínio/química , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Domínio Catalítico , Clonagem Molecular , Cristalografia por Raios X , Distroglicanas/metabolismo , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Fluoretos/química , Expressão Gênica , Humanos , Magnésio/metabolismo , Manose/metabolismo , Modelos Moleculares , Mutação , Fosforilação , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Células Sf9 , Especificidade por Substrato , Trissacarídeos/metabolismo , Peixe-Zebra/metabolismo
19.
Proc Natl Acad Sci U S A ; 113(25): E3482-91, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27185916

RESUMO

The modification of proteins by phosphorylation occurs in all life forms and is catalyzed by a large superfamily of enzymes known as protein kinases. We recently discovered a family of secretory pathway kinases that phosphorylate extracellular proteins. One member, family with sequence similarity 20C (Fam20C), is the physiological Golgi casein kinase. While examining distantly related protein sequences, we observed low levels of identity between the spore coat protein H (CotH), and the Fam20C-related secretory pathway kinases. CotH is a component of the spore in many bacterial and eukaryotic species, and is required for efficient germination of spores in Bacillus subtilis; however, the mechanism by which CotH affects germination is unclear. Here, we show that CotH is a protein kinase. The crystal structure of CotH reveals an atypical protein kinase-like fold with a unique mode of ATP binding. Examination of the genes neighboring cotH in B. subtilis led us to identify two spore coat proteins, CotB and CotG, as CotH substrates. Furthermore, we show that CotH-dependent phosphorylation of CotB and CotG is required for the efficient germination of B. subtilis spores. Collectively, our results define a family of atypical protein kinases and reveal an unexpected role for protein phosphorylation in spore biology.


Assuntos
Proteínas Quinases , Esporos Bacterianos/genética , Bacillus subtilis/genética , Proteínas de Bactérias/química , Proteínas do Capsídeo , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA