Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
1.
J Chem Inf Model ; 63(1): 224-239, 2023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36374995

RESUMO

The intricate mechanisms of allosteric regulation in kinases are of general interest to the scientific community for potential therapeutic implications. However, the diversity among kinases and their regulatory routes requires a case-by-case study to widen the repertoire of known mechanisms. The present study achieves this by understanding TAK1 kinase activation by TAB1 as a model phenomenon for the first time. Despite the known capacity of TAK1 to switch between its inactive ("DFG-out") and active-like ("DFG-in") conformations, the questionable role of TAB1 in offering an energetic favor to this has been addressed here using sequential combination of enhanced sampling methods like targeted molecular dynamics (TMD) and Gaussian accelerated molecular dynamics (GaMD). It reveals how a minimal domain of TAB1 sufficiently acts like a "catalytic gear" by favorably sculpting TAK1's thermodynamic landscape (potential of mean force in 2D) that accelerates "in"-"out" conformational switching of the conserved DFG motif. Standard molecular dynamics simulations (∼5 µs) reveal that TAB1 fascinatingly exploits the "lever-like" αF helix of TAK1 kinase domain to remotely propel the DFG motif via subtle helical "unfolding-folding" modifications within the kinase activation loop. The presence of two charged residues on terminal poles of αF helix imparts it, with this unique "lever-like" utility, and this turns out to be one important signature of co-evolution between TAK1 and TAB1. The entire mechanism of TAB1's impact transduction, which is found to be analogous to the moves in the popular "Chinese checker" game, gives a clear proof of the "dynamics-driven allostery" concept in kinases. The findings further benchmark TAK1's known autophosphorylation capacity. A novel insight into kinase allostery is thus provided, which potentiates investigation of similar capacities in other kinases.


Assuntos
MAP Quinase Quinase Quinases , MAP Quinase Quinase Quinases/química , MAP Quinase Quinase Quinases/metabolismo , Fosforilação
2.
Plant Genome ; 15(3): e20216, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35535627

RESUMO

Mitogen-activated protein kinase (MAPK or MPK) cascades consist of three protein kinase components, MAPK kinase kinases (MAPKKKs), MAPK kinases (MKKs and MPKs), which are indispensable for various plant physiological processes. The functions of MAPK families have been extensively studied in maize (Zea mays L.) and other plant species, but little is known about MAPK families in the elite Chinese maize line Huangzaosi (hzs). In this study, we observed that overall performance of Huangzaosi was substantially better than that of B73 under drought conditions at the seedling and V16 stages with a favorable root/canopy ratio. In silico analyses identified 72, 10, and 24 MAPKKKs, MKKs, and MPKs, respectively, in Huangzaosi. Examinations of phylogenetic relationships among Arabidopsis thaliana (L.) Heynh., rice (Oryza sativa L.), and maize (lines B73 and hzs), gene structures, conserved protein motifs, and chromosomal locations revealed their evolutionary relationships. The basal gene expression levels and tissue specificities of all three MAPK families in hzs reflected the diversity in the MAPK functions related to growth and development. The quantitative real-time polymerase chain reaction (qPCR) assay indicated that certain MAPK genes with high basal expression levels in the primary and crown roots responded differentially to drought between B73 and hzs, suggesting that these genes may contribute to their distinct drought tolerance at different developmental stages. The important information regarding the evolution and expression of hzs MAPK family members generated in this study provides a new avenue for the better understanding on the regulatory mechanism of MAPK cascade in the core inbred line hzs, which may be useful to guide the development of new maize cultivars with desirable traits (e.g., drought resistance).


Assuntos
Oryza , Zea mays , China , MAP Quinase Quinase Quinases/química , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/química , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/química , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Filogenia , Zea mays/genética , Zea mays/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34470822

RESUMO

The RAF/MEK/ERK pathway is central to the control of cell physiology, and its dysregulation is associated with many cancers. Accordingly, the proteins constituting this pathway, including MEK1/2 (MEK), have been subject to intense drug discovery and development efforts. Allosteric MEK inhibitors (MEKi) exert complex effects on RAF/MEK/ERK pathway signaling and are employed clinically in combination with BRAF inhibitors in malignant melanoma. Although mechanisms and structures of MEKi bound to MEK have been described for many of these compounds, recent studies suggest that RAF/MEK complexes, rather than free MEK, should be evaluated as the target of MEKi. Here, we describe structural and biochemical studies of eight structurally diverse, clinical-stage MEKi to better understand their mechanism of action on BRAF/MEK complexes. We find that all of these agents bind in the MEK allosteric site in BRAF/MEK complexes, in which they stabilize the MEK activation loop in a conformation that is resistant to BRAF-mediated dual phosphorylation required for full activation of MEK. We also show that allosteric MEK inhibitors act most potently on BRAF/MEK complexes rather than on free active MEK, further supporting the notion that a BRAF/MEK complex is the physiologically relevant pharmacologic target for this class of compounds. Our findings provide a conceptual and structural framework for rational development of RAF-selective MEK inhibitors as an avenue to more effective and better-tolerated agents targeting this pathway.


Assuntos
MAP Quinase Quinase Quinases/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Regulação Alostérica , Ativação Enzimática , Estabilidade Enzimática , Humanos , MAP Quinase Quinase Quinases/química , MAP Quinase Quinase Quinases/metabolismo , Fosforilação , Conformação Proteica , Transdução de Sinais
4.
Nat Commun ; 12(1): 1353, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33649309

RESUMO

Cells are under threat of osmotic perturbation; cell volume maintenance is critical in cerebral edema, inflammation and aging, in which prominent changes in intracellular or extracellular osmolality emerge. After osmotic stress-enforced cell swelling or shrinkage, the cells regulate intracellular osmolality to recover their volume. However, the mechanisms recognizing osmotic stress remain obscured. We previously clarified that apoptosis signal-regulating kinase 3 (ASK3) bidirectionally responds to osmotic stress and regulates cell volume recovery. Here, we show that macromolecular crowding induces liquid-demixing condensates of ASK3 under hyperosmotic stress, which transduce osmosensing signal into ASK3 inactivation. A genome-wide small interfering RNA (siRNA) screen identifies an ASK3 inactivation regulator, nicotinamide phosphoribosyltransferase (NAMPT), related to poly(ADP-ribose) signaling. Furthermore, we clarify that poly(ADP-ribose) keeps ASK3 condensates in the liquid phase and enables ASK3 to become inactivated under hyperosmotic stress. Our findings demonstrate that cells rationally incorporate physicochemical phase separation into their osmosensing systems.


Assuntos
Lubrificação , Pressão Osmótica , Poli Adenosina Difosfato Ribose/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Citocinas/metabolismo , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/química , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , MAP Quinase Quinase Quinases/ultraestrutura , Modelos Moleculares , Mutação/genética , NAD/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Domínios Proteicos
5.
Org Biomol Chem ; 19(6): 1412-1425, 2021 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-33501482

RESUMO

TAK1 is a serine/threonine kinase which is involved in the moderation of cell survival and death via the TNFα signalling pathway. It is also implicated in a range of cancer and anti-inflammatory diseases. Drug discovery efforts on this target have focused on both traditional reversible ATP-binding site inhibitors and increasingly popular irreversible covalent binding inhibitors. Irreversible inhibitors can offer benefits in terms of potency, selectivity and PK/PD meaning they are increasingly pursued where the strategy exists. TAK1 kinase differs from the better-known kinase EGFR in that the reactive cysteine nucleophile targeted by electrophilic inhibitors is located towards the back of the ATP binding site, not at its mouth. While a wealth of structural and computational effort has been spent exploring EGFR, only limited studies on TAK1 have been reported. In this work we report the first QM/MM study on TAK1 aiming to better understand aspects of covalent adduct formation. Our goal is to identify the general base in the catalytic reaction, whether the process proceeds via a stepwise or concerted pathway, and how the highly flexible G-loop and A-loop affect the catalytic cysteine located nearby.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Domínio Catalítico , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/química , Simulação de Dinâmica Molecular , Ligação Proteica , Inibidores de Proteínas Quinases/química , Teoria Quântica
6.
Open Biol ; 10(9): 200099, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32873150

RESUMO

Aberrant tumour necrosis factor (TNF) signalling is a hallmark of many inflammatory diseases including rheumatoid arthritis (RA), irritable bowel disease and lupus. Maladaptive TNF signalling can lead to hyper active downstream nuclear factor (NF)-κß signalling in turn amplifying a cell's inflammatory response and exacerbating disease. Within the TNF intracellular inflammatory signalling cascade, transforming growth factor-ß-activated kinase 1 (TAK1) has been shown to play a critical role in mediating signal transduction and downstream NF-κß activation. Owing to its role in TNF inflammatory signalling, TAK1 has become a potential therapeutic target for the treatment of inflammatory diseases such as RA. This review highlights the current development of targeting the TNF-TAK1 signalling axis as a novel therapeutic strategy for the treatment of inflammatory diseases.


Assuntos
Inflamação/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Desenvolvimento de Medicamentos/métodos , Humanos , Inflamação/tratamento farmacológico , Inflamação/etiologia , Mediadores da Inflamação/metabolismo , MAP Quinase Quinase Quinases/química , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Inibidores do Fator de Necrose Tumoral/química , Inibidores do Fator de Necrose Tumoral/farmacologia , Inibidores do Fator de Necrose Tumoral/uso terapêutico , Fator de Necrose Tumoral alfa/química
7.
Biochemistry ; 59(31): 2849-2858, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32667811

RESUMO

The sterile α motif, also called the SAM domain, is known to form homo or heterocomplexes that modulate diverse biological functions through the regulation of specific protein-protein interactions. The MAPK pathway of budding yeast Saccharomyces cerevisiae is comprised of a three-tier kinase system akin to mammals. The MAPKKK Ste11 protein of yeast contains a homodimer SAM domain, which is critical for transmitting cues to the downstream kinases. The structural stability of the dimeric Ste11 SAM is maintained by hydrophobic and ionic interactions at the interfacial amino acids. The urea-induced equilibrium-unfolding process of the Ste11 SAM domain is cooperative without evidence of any intermediate states. The native-state H/D exchange under subdenaturing conditions is a useful method for the detection of intermediate states of proteins. In the present study, we investigated the effect of ionic strength on the conformational stability of the dimer using the H/D exchange experiments. The hydrogen exchange behavior of the Ste11 dimer under physiological salt concentrations reveals two partially unfolded metastable intermediate states, which may be generated by a sequential and cooperative unfolding of the five helices present in the domain. These intermediates appear to be significant for the reversible unfolding kinetics via hydrophobic collapse. In contrast, higher ionic concentrations eliminate this cooperative interactions that stabilize the pairs of helices.


Assuntos
Medição da Troca de Deutério , MAP Quinase Quinase Quinases/química , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Multimerização Proteica/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/química , Cloreto de Sódio/farmacologia , Relação Dose-Resposta a Droga , Estabilidade Enzimática/efeitos dos fármacos , Espectrometria de Massa com Troca Hidrogênio-Deutério , Estrutura Quaternária de Proteína , Desdobramento de Proteína/efeitos dos fármacos , Ureia/farmacologia
9.
Sci Signal ; 13(622)2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32156783

RESUMO

Apoptosis signal-regulating kinases (ASK1, ASK2, and ASK3) are activators of the p38 and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinase (MAPK) pathways. ASK1-3 form oligomeric complexes known as ASK signalosomes that initiate signaling cascades in response to diverse stress stimuli. Here, we demonstrated that oligomerization of ASK proteins is driven by previously uncharacterized sterile-alpha motif (SAM) domains that reside at the carboxy-terminus of each ASK protein. SAM domains from ASK1-3 exhibited distinct behaviors, with the SAM domain of ASK1 forming unstable oligomers, that of ASK2 remaining predominantly monomeric, and that of ASK3 forming a stable oligomer even at a low concentration. In contrast to their behavior in isolation, the ASK1 and ASK2 SAM domains preferentially formed a stable heterocomplex. The crystal structure of the ASK3 SAM domain, small-angle x-ray scattering, and mutagenesis suggested that ASK3 oligomers and ASK1-ASK2 complexes formed discrete, quasi-helical rings through interactions between the mid-loop of one molecule and the end helix of another molecule. Preferential ASK1-ASK2 binding was consistent with mass spectrometry showing that full-length ASK1 formed hetero-oligomeric complexes incorporating large amounts of ASK2. Accordingly, disrupting the association between SAM domains impaired ASK activity in the context of electrophilic stress induced by 4-hydroxy-2-nonenal (HNE). These findings provide a structural template for how ASK proteins assemble foci that drive inflammatory signaling and reinforce the notion that strategies to target ASK proteins should consider the concerted actions of multiple ASK family members.


Assuntos
MAP Quinase Quinase Quinase 5/química , MAP Quinase Quinase Quinases/química , Complexos Multienzimáticos/química , Multimerização Proteica , Células HEK293 , Humanos , MAP Quinase Quinase Quinase 5/genética , MAP Quinase Quinase Quinase 5/metabolismo , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Domínios Proteicos
10.
Biomed Pharmacother ; 126: 110084, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32171166

RESUMO

Sepsis is a main reason for death in intensive care units, inflammation is closely related to sepsis. Anti-inflammation plays an important role in treating of sepsis. ZT01 is a triptolide derivative with strong anti-inflammatory activity and low toxicity. The purpose of this study is to evaluate the anti-inflammatory activity of ZT01 under the sepsis condition and explore the underlying molecular mechanisms. Two in vivo model of sepsis, caecal ligation and puncture or intraperitoneal injection of LPS in C57BL/6, were used to evaluate the therapeutic effects of ZT01. In vitro, the anti-inflammatory properties of ZT01 were assessed in IFN-γ or LPS-induced macrophages by ELISA, RT-PCR, western blotting and co-immunoprecipitation. Macrophages were used to investigate the polarization phenotype by flow cytometry. The results showed, ZT01 significantly attenuated inflammatory response of sepsis in serum or lung tissue by inhibiting production of pro-inflammatory factors and improved the survival rate of septic mice in vivo. In cultured macrophages, ZT01 not only decreased the levels of TNF-α and IL-6 but also prevented the TKA1-TAB1 complex formation, thereby inhibiting the phosphorylation expression of MKK4 and JNK, which were all stimulated by LPS. Moreover, ZT01 inhibited the LPS-induced polarization of macrophages into pro-inflammatory phenotype. Adoptive transfer ZT01 pretreated bone marrow-derived macrophages obviously reduced the pro-inflammatory factors in mice after LPS challenge. Our findings suggested that ZT01 exhibited anti-inflammation activity via preventing the pro-inflammatory phenotype of macrophages by blocking the formation of the TAK1-TAB1 complex and subsequently phosphorylation of MKK4 and JNK.


Assuntos
Anti-Inflamatórios/farmacologia , Diterpenos/farmacologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Fenantrenos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Animais , Anti-Inflamatórios/química , Células Cultivadas , Citocinas/metabolismo , Diterpenos/química , Compostos de Epóxi/química , Compostos de Epóxi/farmacologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/efeitos adversos , MAP Quinase Quinase Quinases/química , Macrófagos/metabolismo , Masculino , Camundongos , Modelos Biológicos , Modelos Moleculares , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Fenantrenos/química , Fosforilação , Ligação Proteica , Inibidores de Proteínas Quinases/química , Células RAW 264.7 , Sepse/tratamento farmacológico , Sepse/etiologia , Sepse/metabolismo , Sepse/mortalidade , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
11.
J Med Chem ; 63(5): 2114-2130, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-31244114

RESUMO

ZAK is a new promising target for discovery of drugs with activity against antihypertrophic cardiomyopathy (HCM). A series of 1,2,3-triazole benzenesulfonamides were designed and synthesized as selective ZAK inhibitors. One of these compounds, 6p binds tightly to ZAK protein (Kd = 8.0 nM) and potently suppresses the kinase function of ZAK with single-digit nM (IC50 = 4.0 nM) and exhibits excellent selectivity in a KINOMEscan screening platform against a panel of 403 wild-type kinases. This compound dose dependently blocks p38/GATA-4 and JNK/c-Jun signaling and demonstrates promising in vivo anti-HCM efficacy upon oral administration in a spontaneous hypertensive rat (SHR) model. Compound 6p may serve as a lead compound for new anti-HCM drug discovery.


Assuntos
Desenho de Fármacos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Sulfonamidas/farmacologia , Triazóis/farmacologia , Animais , Humanos , Zíper de Leucina/efeitos dos fármacos , MAP Quinase Quinase Quinases/química , MAP Quinase Quinase Quinases/metabolismo , Simulação de Acoplamento Molecular , Inibidores de Proteínas Quinases/sangue , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Relação Estrutura-Atividade , Sulfonamidas/sangue , Sulfonamidas/síntese química , Sulfonamidas/química , Triazóis/sangue , Triazóis/síntese química , Triazóis/química , Benzenossulfonamidas
12.
Fish Shellfish Immunol ; 94: 264-270, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31499204

RESUMO

Transforming growth factor-ß activated kinase-1 (TAK1) is an important member of the mitogen-activated protein kinase kinase kinase (MAP3K) family, which plays an important role in animal innate immune response. However, the TAK1 gene has yet not been reported in amphioxus to date. Here, we have identified and characterized a TAK1 gene from amphioxus (Branchiostoma belcheri) (named as AmphiTAK1) with the full-length cDNA of 3479 bp, including an ORF sequence of 1905 bp, a 5' UTR of 394 bp and a 3' UTR of 1180 bp. Moreover, the predicted AmphiTAK1 protein contains STKc_TAK1 domain, TAB1 and TAB2/3 binding domain which are conserved among chordate, and phylogenetic analysis also shows that the AmphiTAK1 is located at the bottom of the chordate, revealing AmphiTAK1 as a new member of the TAK1 gene family. The further qRT-PCR analysis has shown that AmphiTAK1 is widely expressed in six investigated tissues (gonad, gill, hepatic cecum, intestine, muscle and notochord) of Branchiostoma belcheri, with high expression in notochord and gonad, moderate in gill and hepatic cecum. Notably, the expression level of AmphiTAK1 is significantly up-regulated after LPS stimulation. Specially, we also find that AmphiTAK1 protein can interact with AmphiTAB1 by immunoprecipitation assay. These findings reveal that AmphiTAK1 might interact with AmphiTAB1 to involve in innate immune response of Branchiostoma belcheri. Taken together, our present works provide a new insight into evolution and innate immune response mechanism of AmphiTAK1 gene in Branchiostoma belcheri.


Assuntos
Regulação da Expressão Gênica/imunologia , Imunidade Inata/genética , Anfioxos/genética , Anfioxos/imunologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/imunologia , Sequência de Aminoácidos , Animais , Evolução Molecular , Perfilação da Expressão Gênica , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase Quinases/química , Filogenia , Alinhamento de Sequência
13.
Mol Cells ; 42(9): 646-660, 2019 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-31480825

RESUMO

Abscisic acid (ABA) is a phytohormone essential for seed development and seedling growth under unfavorable environmental conditions. The signaling pathway leading to ABA response has been established, but relatively little is known about the functional regulation of the constituent signaling components. Here, we present several lines of evidence that Arabidopsis Raf-like kinase Raf10 modulates the core ABA signaling downstream of signal perception step. In particular, Raf10 phosphorylates subclass III SnRK2s (SnRK2.2, SnRK2.3, and SnRK2.6), which are key positive regulators, and our study focused on SnRK2.3 indicates that Raf10 enhances its kinase activity and may facilitate its release from negative regulators. Raf10 also phosphorylates transcription factors (ABI5, ABF2, and ABI3) critical for ABAregulted gene expression. Furthermore, Raf10 was found to be essential for the in vivo functions of SnRK2s and ABI5. Collectively, our data demonstrate that Raf10 is a novel regulatory component of core ABA signaling.


Assuntos
Ácido Abscísico/metabolismo , Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Proteínas de Arabidopsis/química , MAP Quinase Quinase Quinases/química , Fenótipo , Fosforilação , Fosfosserina/metabolismo , Fosfotreonina/metabolismo , Multimerização Proteica
14.
Genome ; 62(9): 609-622, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31271725

RESUMO

Mitogen-activated protein kinase kinase kinases (MAPKKKs) are important components of MAPK cascades, which have different functions during developmental processes and stress responses. To date, there has been no systematic investigation of this gene family in the diploid cotton Gossypium arboreum L. In this study, a genome-wide survey was performed that identified 78 MAPKKK genes in G. arboreum. Phylogenetic analysis classified these genes into three subgroups: 14 belonged to ZIK, 20 to MEKK, and 44 to Raf. Chromosome location, phylogeny, and the conserved protein motifs of the MAPKKK gene family in G. arboreum were analyzed. The MAPKKK genes had a scattered genomic distribution across 13 chromosomes. The members in the same subfamily shared similar conserved motifs. The MAPKKK expression patterns were analyzed in mature leaves, stems, roots, and at different ovule developmental stages, as well as under salt and drought stresses. Transcriptome analysis showed that 76 MAPKKK genes had different transcript accumulation patterns in the tested tissues and 38 MAPKKK genes were differentially expressed in response to salt and drought stresses. These results lay the foundation for understanding the complex mechanisms behind MAPKKK-mediated developmental processes and abiotic stress-signaling transduction pathways in cotton.


Assuntos
Gossypium/genética , MAP Quinase Quinase Quinases/genética , Proteínas de Plantas/genética , Motivos de Aminoácidos , Mapeamento Cromossômico , Cromossomos de Plantas , Perfilação da Expressão Gênica , Gossypium/enzimologia , MAP Quinase Quinase Quinases/química , Família Multigênica , Filogenia , Proteínas de Plantas/química
15.
Fish Shellfish Immunol ; 90: 80-90, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31022453

RESUMO

Transforming growth factor-ß-activating kinase 1 (TAK1) is essential for diverse important biological functions, such as innate immunity, development and cell survival. In the present study, the homologs of TAK1 and TAK1-binding protein 1 (TAB1) were identified and characterized from mud crab Scylla paramamosain for the first time. The full-length cDNAs of SpTAK1 and SpTAB1 were 2, 226 bp and 2, 433 bp with 1, 782 bp and 1, 533 bp open reading frame (ORF), respectively. The deduced SpTAK1 protein contained a conserved S_TKc (Serine/threonine protein kinases, catalytic) domain, and the putative SpTAB1 protein possessed a typical PP2Cc (Serine/threonine phosphatases, family 2C, catalytic) domain and a potential TAK1 docking motif. Real-time PCR analysis showed that SpTAK1 and SpTAB1 were highly expressed at early development stages, suggesting their participation in crab's development process. Moreover, the expression levels of SpTAK1 and SpTAB1 in hepatopancreas were positively stimulated after challenge with Vibro alginolyticus and Poly (I:C), implying the involvement of SpTAK1 and SpTAB1 in innate immune responses against both bacterial and viral infections. When SpTAK1 or SpTAB1 were silenced in vivo, the expression levels of two IMDNFκB signaling components (SpIKKß and SpRelish) and six antimicrobial peptide (AMP) genes (SpALF1-5 and SpCrustin) were significantly reduced, and the bacteria clearance capacity of crabs was also markedly impaired in SpTAK1 or SpTAB1 silenced crabs. Additionally, overexpression of SpTAK1 and SpTAB1 in HEK293T cells could markedly activate the mammalian NF-κB signaling. Collectively, our results suggested that TAK1 and TAB1 regulated crab's innate immunity via modulating the IMDNFκB signaling. These findings may provide new insights into the TAK1/TAB1-mediated signaling cascades in crustaceans and pave the way for a better understanding of crustacean innate immune system.


Assuntos
Proteínas de Artrópodes/genética , Proteínas de Artrópodes/imunologia , Braquiúros/genética , Braquiúros/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Artrópodes/química , Perfilação da Expressão Gênica , MAP Quinase Quinase Quinases/química , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/imunologia , Filogenia , Alinhamento de Sequência , Transdução de Sinais/genética
16.
Mol Plant Pathol ; 19(12): 2590-2602, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30047240

RESUMO

Many obligately parasitic pathogens absorb nutrients from host plants via specialized infection structures, called haustoria and infection hyphae, to further colonization and growth in the host plant. In the wheat (Triticum aestivum) stripe rust fungus, Puccinia striiformis f. sp. tritici (Pst), the mitogen-activated protein kinase kinase (MAPKK) PsFUZ7 is involved in the regulation of haustorium formation and invasive growth. Here, we functionally characterized PsKPP4 of Pst, which is homologous to the yeast MAPKKK STE11. Similar to the silencing of PsFUZ7, the knockdown of PsKPP4 was detected in the vegetative hyphae and haustoria, resulting in the reduced pathogenicity of Pst. Pst urediniospores treated with the STE11 MAPKKK activation inhibitor produced deformed germ tubes. In addition, overexpression of PsKPP4 in fission yeast resulted in the production of fusiform cells and increased tolerance of yeast cells to oxidative stress. The transformation of PsKPP4 into the mst11 mutant of Magnaporthe oryzae partially restored mst11 function. The PsKPP4 protein contains a sterile alpha motif (SAM), Ras association (RA) and kinase domains, similar to its homologues in other fungi. Yeast two-hybrid assays revealed that the SAM domain is essential for the interaction between PsKPP4 and PsUBC2, a homologue of Ustilago maydis UBC2, known to interact with KPP4, which is associated with the regulation of the Fus3 cascade. Host-induced gene silencing of PsUBC2 reduced the pathogenicity of Pst slightly, indicating that PsUBC2 also plays a minor role in the regulation of the infection pathway of Pst. These observations indicate that PsKPP4, interacting with PsUBC2, may play an important role in the regulation of infection-related morphogenesis in Pst.


Assuntos
Basidiomycota/genética , Basidiomycota/patogenicidade , Proteínas Fúngicas/genética , Inativação Gênica , Genes Fúngicos , MAP Quinase Quinase Quinases/genética , Doenças das Plantas/genética , Doenças das Plantas/microbiologia , Sequência de Aminoácidos , Basidiomycota/crescimento & desenvolvimento , Sequência Conservada , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Genes Supressores , Hifas/crescimento & desenvolvimento , MAP Quinase Quinase Quinases/química , Magnaporthe/fisiologia , Mutação/genética , Domínios Proteicos , Schizosaccharomyces/metabolismo , Estresse Fisiológico
17.
J Biol Chem ; 293(35): 13553-13565, 2018 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-29980598

RESUMO

Mixed-lineage kinase 3 (MLK3; also known as MAP3K11) is a Ser/Thr protein kinase widely expressed in normal and cancerous tissues, including brain, lung, liver, heart, and skeletal muscle tissues. Its Src homology 3 (SH3) domain has been implicated in MLK3 autoinhibition and interactions with other proteins, including those from viruses. The MLK3 SH3 domain contains a six-amino-acid insert corresponding to the n-Src insert, suggesting that MLK3 may bind additional peptides. Here, affinity selection of a phage-displayed combinatorial peptide library for MLK3's SH3 domain yielded a 13-mer peptide, designated "MLK3 SH3-interacting peptide" (MIP). Unlike most SH3 domain peptide ligands, MIP contained a single proline. The 1.2-Å crystal structure of the MIP-bound SH3 domain revealed that the peptide adopts a ß-hairpin shape, and comparison with a 1.5-Å apo SH3 domain structure disclosed that the n-Src loop in SH3 undergoes an MIP-induced conformational change. A 1.5-Å structure of the MLK3 SH3 domain bound to a canonical proline-rich peptide from hepatitis C virus nonstructural 5A (NS5A) protein revealed that it and MIP bind the SH3 domain at two distinct sites, but biophysical analyses suggested that the two peptides compete with each other for SH3 binding. Moreover, SH3 domains of MLK1 and MLK4, but not MLK2, also bound MIP, suggesting that the MLK1-4 family may be differentially regulated through their SH3 domains. In summary, we have identified two distinct peptide-binding sites in the SH3 domain of MLK3, providing critical insights into mechanisms of ligand binding by the MLK family of kinases.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Humanos , MAP Quinase Quinase Quinases/química , Simulação de Acoplamento Molecular , Biblioteca de Peptídeos , Peptídeos/química , Ligação Proteica , Domínios de Homologia de src , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
18.
Nat Med ; 24(2): 213-223, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29291351

RESUMO

Nonalcoholic steatohepatitis (NASH) is a common clinical condition that can lead to advanced liver diseases. Lack of effective pharmacotherapies for NASH is largely attributable to an incomplete understanding of its pathogenesis. The deubiquitinase cylindromatosis (CYLD) plays key roles in inflammation and cancer. Here we identified CYLD as a suppressor of NASH in mice and in monkeys. CYLD is progressively degraded upon interaction with the E3 ligase TRIM47 in proportion to NASH severity. We observed that overexpression of Cyld in hepatocytes concomitantly inhibits lipid accumulation, insulin resistance, inflammation and fibrosis in mice with NASH induced in an experimental setting. Mechanistically, CYLD interacts directly with the kinase TAK1 and removes its K63-linked polyubiquitin chain, which blocks downstream activation of the JNK-p38 cascades. Notably, reconstitution of hepatic CYLD expression effectively reverses disease progression in mice with dietary or genetically induced NASH and in high-fat diet-fed monkeys predisposed to metabolic syndrome. Collectively, our findings demonstrate that CYLD mitigates NASH severity and identify the CYLD-TAK1 axis as a promising therapeutic target for management of the disease.


Assuntos
Cisteína Endopeptidases/genética , Inflamação/genética , MAP Quinase Quinase Quinases/genética , Hepatopatia Gordurosa não Alcoólica/genética , Animais , Proteínas de Transporte/genética , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Enzima Desubiquitinante CYLD , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Haplorrinos , Humanos , Inflamação/fisiopatologia , Fígado/metabolismo , Fígado/patologia , MAP Quinase Quinase 4/genética , MAP Quinase Quinase Quinases/química , MAP Quinase Quinase Quinases/metabolismo , Síndrome Metabólica/genética , Síndrome Metabólica/patologia , Camundongos , Proteínas de Neoplasias/genética , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Proteínas Nucleares/genética , Ligação Proteica/genética , Índice de Gravidade de Doença , Transdução de Sinais/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
19.
J Biol Chem ; 293(8): 2661-2674, 2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29196608

RESUMO

TRIM5α is an important host restriction factor that could potently block retrovirus infection. The SPRY domain of TRIM5α mediates post-entry restriction by recognition of and binding to the retroviral capsid. Human TRIM5α also functions as an innate immune sensor to activate AP-1 and NF-κB signaling, which subsequently restrict virus replication. Previous studies have shown that the AP-1 and NF-κB signaling activation relies on the RING motif of TRIM5α. In this study, we have demonstrated that the SPRY domain is essential for rhesus macaque TRIM5α to activate AP-1 but not NF-κB signaling. The AP-1 activation mainly depends on all of the ß-sheet barrel on SPRY structure of TRIM5α. Furthermore, the SPRY-mediated auto-ubiquitination of TRIM5α is required for AP-1 activation. This study reports that rhesus macaque TRIM5α mainly undergoes Lys27-linked and Met1-linked auto-polyubiquitination. Finally, we found that the TRIM5α signaling function was positively correlated with its retroviral restriction activity. This study discovered an important role of the SPRY domain in immune signaling and antiviral activity and further expanded our knowledge of the antiviral mechanism of TRIM5α.


Assuntos
Domínio B30.2-SPRY , Modelos Moleculares , Proteína de Replicação C/metabolismo , Transdução de Sinais , Proteínas com Motivo Tripartido/metabolismo , Ubiquitinação , Animais , Ativação Enzimática , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células HeLa , Humanos , MAP Quinase Quinase Quinases/química , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Macaca fascicularis , Macaca mulatta , NF-kappa B/agonistas , NF-kappa B/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica em Folha beta , Domínios RING Finger , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteína de Replicação C/química , Proteína de Replicação C/genética , Especificidade da Espécie , Proteínas com Motivo Tripartido/química , Proteínas com Motivo Tripartido/genética
20.
Pharmacol Res ; 129: 188-193, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29183769

RESUMO

Tumor progression locus 2 (TPL2, also known as COT or MAP3K8) is a mitogen-activated protein kinase kinase (MAP3K) activated downstream of TNFαR, IL1R, TLR, CD40, IL17R, and some GPCRs. TPL2 regulates the MEK1/2 and ERK1/2 pathways to regulate a cascade of inflammatory responses. In parallel to this, TPL2 also activates p38α and p38δ to drive the production of various inflammatory mediators in neutrophils. We discuss the implications of this finding in the context of various inflammatory diseases.


Assuntos
Inflamação/metabolismo , MAP Quinase Quinase Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Autoimunidade , Humanos , MAP Quinase Quinase Quinases/química , Proteínas Proto-Oncogênicas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...