Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Gastroenterol Hepatol ; 17(6): 983-1005, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38307491

RESUMO

Microvillus inclusion disease (MVID) is a rare condition that is present from birth and affects the digestive system. People with MVID experience severe diarrhea that is difficult to control, cannot absorb dietary nutrients, and struggle to grow and thrive. In addition, diverse clinical manifestations, some of which are life-threatening, have been reported in cases of MVID. MVID can be caused by variants in the MYO5B, STX3, STXBP2, or UNC45A gene. These genes produce proteins that have been functionally linked to each other in intestinal epithelial cells. MVID associated with STXBP2 variants presents in a subset of patients diagnosed with familial hemophagocytic lymphohistiocytosis type 5. MVID associated with UNC45A variants presents in most patients diagnosed with osteo-oto-hepato-enteric syndrome. Furthermore, variants in MYO5B or STX3 can also cause other diseases that are characterized by phenotypes that can co-occur in subsets of patients diagnosed with MVID. Recent studies involving clinical data and experiments with cells and animals revealed connections between specific phenotypes occurring outside of the digestive system and the type of gene variants that cause MVID. Here, we have reviewed these patterns and correlations, which are expected to be valuable for healthcare professionals in managing the disease and providing personalized care for patients and their families.


Assuntos
Síndromes de Malabsorção , Microvilosidades , Mucolipidoses , Fenótipo , Humanos , Mucolipidoses/genética , Mucolipidoses/patologia , Microvilosidades/patologia , Microvilosidades/genética , Síndromes de Malabsorção/genética , Síndromes de Malabsorção/patologia , Animais , Miosina Tipo V/genética , Miosina Tipo V/metabolismo , Mutação , Predisposição Genética para Doença
2.
Dev Cell ; 58(22): 2477-2494.e8, 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37875118

RESUMO

Cilia protrude from the cell surface and play critical roles in intracellular signaling, environmental sensing, and development. Reduced actin-dependent contractility and intracellular trafficking are both required for ciliogenesis, but little is known about how these processes are coordinated. Here, we identified a Rac1- and Rab35-binding protein with a truncated BAR (Bin/amphiphysin/Rvs) domain that we named MiniBAR (also known as KIAA0355/GARRE1), which plays a key role in ciliogenesis. MiniBAR colocalizes with Rac1 and Rab35 at the plasma membrane and on intracellular vesicles trafficking to the ciliary base and exhibits fast pulses at the ciliary membrane. MiniBAR depletion leads to short cilia, resulting from abnormal Rac-GTP/Rho-GTP levels and increased acto-myosin-II-dependent contractility together with defective trafficking of IFT88 and ARL13B into cilia. MiniBAR-depleted zebrafish embryos display dysfunctional short cilia and hallmarks of ciliopathies, including left-right asymmetry defects. Thus, MiniBAR is a dual Rac and Rab effector that controls both actin cytoskeleton and membrane trafficking for ciliogenesis.


Assuntos
Proteínas do Citoesqueleto , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Proteínas do Citoesqueleto/metabolismo , Transdução de Sinais , Proteínas de Transporte/metabolismo , Cílios/metabolismo , Guanosina Trifosfato/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
3.
Oncogene ; 40(23): 4019-4032, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34012098

RESUMO

Membrane Type 1 Matrix Metalloprotease (MT1-MMP) contributes to the invasive progression of breast cancers by degrading extracellular matrix tissues. Nucleoside diphosphate kinase, NME1/NM23-H1, has been identified as a metastasis suppressor; however, its contribution to local invasion in breast cancer is not known. Here, we report that NME1 is up-regulated in ductal carcinoma in situ (DCIS) as compared to normal breast epithelial tissues. NME1 levels drop in microinvasive and invasive components of breast tumor cells relative to synchronous DCIS foci. We find a strong anti-correlation between NME1 and plasma membrane MT1-MMP levels in the invasive components of breast tumors, particularly in aggressive histological grade III and triple-negative breast cancers. Knockout of NME1 accelerates the invasive transition of breast tumors in the intraductal xenograft model. At the mechanistic level, we find that MT1-MMP, NME1 and dynamin-2, a GTPase known to require GTP production by NME1 for its membrane fission activity in the endocytic pathway, interact in clathrin-coated vesicles at the plasma membrane. Loss of NME1 function increases MT1-MMP surface levels by inhibiting endocytic clearance. As a consequence, the ECM degradation and invasive potentials of breast cancer cells are enhanced. This study identifies the down-modulation of NME1 as a potent driver of the in situ-to invasive transition during breast cancer progression.


Assuntos
Neoplasias da Mama/metabolismo , Dinamina II/metabolismo , Matriz Extracelular/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular , Movimento Celular/fisiologia , Feminino , Humanos , Metaloproteinase 14 da Matriz/genética , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Metástase Neoplásica , Estadiamento de Neoplasias , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Chem Rev ; 120(1): 5-35, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31689091

RESUMO

Generating force and movement is essential for the functions of cells and organisms. A variety of molecular motors that can move on tracks within cells have evolved to serve this role. How these motors interact with their tracks and how that, in turn, leads to the generation of force and movement is key to understanding the cellular roles that these motor-track systems serve. This review is focused on the best understood of these systems, which is the molecular motor myosin that moves on tracks of filamentous (F-) actin. The review highlights both the progress and the limits of our current understanding of how force generation can be controlled by F-actin-myosin interactions. What has emerged are insights they may serve as a framework for understanding the design principles of a number of types of molecular motors and their interactions with their tracks.


Assuntos
Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/metabolismo , Miosinas/química , Miosinas/metabolismo , Actinas/química , Actinas/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Humanos , Fenômenos Mecânicos , Modelos Moleculares , Domínios Proteicos
5.
Sci Rep ; 9(1): 16036, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31690808

RESUMO

JIP3 and JIP4 (JNK-interacting proteins 3 and 4) are adaptors for cargo recruitment by dynein/dynactin and kinesin1 motors. Both are dimers that are stabilised by two sections of leucine zipper coiled coils. The N-terminal Leucine Zipper I (LZI) belongs to a section that binds dynein-DLIC and kinesin1-KHC, whilst the medial Leucine Zipper II (LZII) binds dynactin-p150glued and kinesin1-KLC. Structural data is available for the LZII, but the LZI section is still uncharacterized. Here we characterize the N-terminal part of JIP3/4 which consists of an RH1 (RILP homology 1) domain followed by the LZI coiled coil using bioinformatical, biophysical and structural approaches. The RH1-LZI tandem of JIP3 associates as a high affinity homodimer exhibiting elongated alpha-helical fold. 3D homology modelling of the RH1-LZI tandem reveals that the kinesin1-KHC binding site mainly overlaps with the RH1 domain. A sequence comparison search indicates that only one other protein family has RH1 domains similar to those of JIP3/4, the RILP (Rab-interacting lysosomal protein) family which consists of adaptor proteins linking Rab GTPases to cytoskeletal motors. RILPL2 is recruited through its RH1 domain by the myosin 5a motor. Here, we showed that the RH1 domain of JIP3 also interacts with myosin 5 A in vitro, highlighting JIP3/4 as possible myosin 5a adaptors. Finally, we propose that JIP3/4 and RILP family members define a unique RH1/RH2-architecture adaptor superfamily linking cytoskeletal motors and Rab GTPases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Citoesqueleto/química , Proteínas do Tecido Nervoso/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Humanos , Zíper de Leucina , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/química , Miosina Tipo V/genética , Miosina Tipo V/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Domínios Proteicos
6.
Small GTPases ; 9(1-2): 22-48, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-28632484

RESUMO

Rab molecular switches are key players in defining membrane identity and regulating intracellular trafficking events in eukaryotic cells. In spite of their global structural similarity, Rab-family members acquired particular features that allow them to perform specific cellular functions. The overall fold and local sequence conservations enable them to utilize a common machinery for prenylation and recycling; while individual Rab structural differences determine interactions with specific partners such as GEFs, GAPs and effector proteins. These interactions orchestrate the spatiotemporal regulation of Rab localization and their turning ON and OFF, leading to tightly controlled Rab-specific functionalities such as membrane composition modifications, recruitment of molecular motors for intracellular trafficking, or recruitment of scaffold proteins that mediate interactions with downstream partners, as well as actin cytoskeleton regulation. In this review we summarize structural information on Rab GTPases and their complexes with protein partners in the context of partner binding specificity and functional outcomes of their interactions in the cell.


Assuntos
Proteínas rab de Ligação ao GTP/química , Proteínas rab de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Animais , Sequência Conservada , Humanos , Ligação Proteica
7.
Hum Mutat ; 39(3): 333-344, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29266534

RESUMO

Microvillus inclusion disease (MVID) is a rare but fatal autosomal recessive congenital diarrheal disorder caused by MYO5B mutations. In 2013, we launched an open-access registry for MVID patients and their MYO5B mutations (www.mvid-central.org). Since then, additional unique MYO5B mutations have been identified in MVID patients, but also in non-MVID patients. Animal models have been generated that formally prove the causality between MYO5B and MVID. Importantly, mutations in two other genes, STXBP2 and STX3, have since been associated with variants of MVID, shedding new light on the pathogenesis of this congenital diarrheal disorder. Here, we review these additional genes and their mutations. Furthermore, we discuss recent data from cell studies that indicate that the three genes are functionally linked and, therefore, may constitute a common disease mechanism that unifies a subset of phenotypically linked congenital diarrheal disorders. We present new data based on patient material to support this. To congregate existing and future information on MVID geno-/phenotypes, we have updated and expanded the MVID registry to include all currently known MVID-associated gene mutations, their demonstrated or predicted functional consequences, and associated clinical information.


Assuntos
Diarreia/congênito , Diarreia/genética , Predisposição Genética para Doença , Proteínas Munc18/genética , Mutação/genética , Miosina Tipo V/genética , Proteínas Qa-SNARE/genética , Animais , Humanos
8.
Nat Commun ; 8(1): 1254, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29093437

RESUMO

The actin and microtubule cytoskeletons play important roles in Golgi structure and function, but how they are connected remain poorly known. In this study, we investigated whether RAB6 GTPase, a Golgi-associated RAB involved in the regulation of several transport steps at the Golgi level, and two of its effectors, Myosin IIA and KIF20A participate in the coupling between actin and microtubule cytoskeleton. We have previously shown that RAB6-Myosin IIA interaction is critical for the fission of RAB6-positive transport carriers from Golgi/TGN membranes. Here we show that KIF20A is also involved in the fission process and serves to anchor RAB6 on Golgi/TGN membranes near microtubule nucleating sites. We provide evidence that the fission events occur at a limited number of hotspots sites. Our results suggest that coupling between actin and microtubule cytoskeletons driven by Myosin II and KIF20A ensures the spatial coordination between RAB6-positive vesicles fission from Golgi/TGN membranes and their exit along microtubules.


Assuntos
Complexo de Golgi/metabolismo , Cinesinas/metabolismo , Proteínas Motores Moleculares/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Vesículas Citoplasmáticas/metabolismo , Humanos , Microtúbulos/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Ratos , Rede trans-Golgi/metabolismo
9.
Nat Commun ; 8: 14528, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28230050

RESUMO

Cytokinetic abscission, the terminal step of cell division, crucially depends on the local constriction of ESCRT-III helices after cytoskeleton disassembly. While the microtubules of the intercellular bridge are cut by the ESCRT-associated enzyme Spastin, the mechanism that clears F-actin at the abscission site is unknown. Here we show that oxidation-mediated depolymerization of actin by the redox enzyme MICAL1 is key for ESCRT-III recruitment and successful abscission. MICAL1 is recruited to the abscission site by the Rab35 GTPase through a direct interaction with a flat three-helix domain found in MICAL1 C terminus. Mechanistically, in vitro assays on single actin filaments demonstrate that MICAL1 is activated by Rab35. Moreover, in our experimental conditions, MICAL1 does not act as a severing enzyme, as initially thought, but instead induces F-actin depolymerization from both ends. Our work reveals an unexpected role for oxidoreduction in triggering local actin depolymerization to control a fundamental step of cell division.


Assuntos
Actinas/metabolismo , Citocinese , Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Proteínas com Domínio LIM/química , Proteínas com Domínio LIM/metabolismo , Proteínas dos Microfilamentos , Oxigenases de Função Mista , Modelos Biológicos , Oxirredução , Polimerização , Ligação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Transporte Proteico , Proteínas rab de Ligação ao GTP/metabolismo
10.
Elife ; 52016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27623148

RESUMO

There is growing evidence for a coupling of actin assembly and myosin motor activity in cells. However, mechanisms for recruitment of actin nucleators and motors on specific membrane compartments remain unclear. Here we report how Spir actin nucleators and myosin V motors coordinate their specific membrane recruitment. The myosin V globular tail domain (MyoV-GTD) interacts directly with an evolutionarily conserved Spir sequence motif. We determined crystal structures of MyoVa-GTD bound either to the Spir-2 motif or to Rab11 and show that a Spir-2:MyoVa:Rab11 complex can form. The ternary complex architecture explains how Rab11 vesicles support coordinated F-actin nucleation and myosin force generation for vesicle transport and tethering. New insights are also provided into how myosin activation can be coupled with the generation of actin tracks. Since MyoV binds several Rab GTPases, synchronized nucleator and motor targeting could provide a common mechanism to control force generation and motility in different cellular processes.


Assuntos
Vesículas Citoplasmáticas/metabolismo , Membranas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Miosina Tipo V/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Cristalografia por Raios X , Camundongos , Proteínas dos Microfilamentos/química , Modelos Moleculares , Miosina Tipo V/química , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Proteínas rab de Ligação ao GTP/química
11.
Nat Commun ; 7: 12456, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27580874

RESUMO

Myosin X has features not found in other myosins. Its structure must underlie its unique ability to generate filopodia, which are essential for neuritogenesis, wound healing, cancer metastasis and some pathogenic infections. By determining high-resolution structures of key components of this motor, and characterizing the in vitro behaviour of the native dimer, we identify the features that explain the myosin X dimer behaviour. Single-molecule studies demonstrate that a native myosin X dimer moves on actin bundles with higher velocities and takes larger steps than on single actin filaments. The largest steps on actin bundles are larger than previously reported for artificially dimerized myosin X constructs or any other myosin. Our model and kinetic data explain why these large steps and high velocities can only occur on bundled filaments. Thus, myosin X functions as an antiparallel dimer in cells with a unique geometry optimized for movement on actin bundles.


Assuntos
Citoesqueleto de Actina/metabolismo , Miosinas/metabolismo , Miosinas/ultraestrutura , Pseudópodes/metabolismo , Animais , Bovinos , Membrana Celular/fisiologia , Cristalografia por Raios X , Dimerização , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular
12.
Proc Natl Acad Sci U S A ; 113(13): E1844-52, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26976594

RESUMO

Molecular motors produce force when they interact with their cellular tracks. For myosin motors, the primary force-generating state has MgADP tightly bound, whereas myosin is strongly bound to actin. We have generated an 8-Å cryoEM reconstruction of this state for myosin V and used molecular dynamics flexed fitting for model building. We compare this state to the subsequent state on actin (Rigor). The ADP-bound structure reveals that the actin-binding cleft is closed, even though MgADP is tightly bound. This state is accomplished by a previously unseen conformation of the ß-sheet underlying the nucleotide pocket. The transition from the force-generating ADP state to Rigor requires a 9.5° rotation of the myosin lever arm, coupled to a ß-sheet rearrangement. Thus, the structure reveals the detailed rearrangements underlying myosin force generation as well as the basis of strain-dependent ADP release that is essential for processive myosins, such as myosin V.


Assuntos
Actinas/metabolismo , Difosfato de Adenosina/metabolismo , Miosina Tipo V/química , Miosina Tipo V/metabolismo , Actinas/química , Sítios de Ligação , Microscopia Crioeletrônica , Cristalografia por Raios X , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Conformação Proteica
13.
Proc Natl Acad Sci U S A ; 112(11): E1201-9, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25751888

RESUMO

Mutations in the reverse-direction myosin, myosin VI, are associated with deafness in humans and mice. A myosin VI deafness mutation, D179Y, which is in the transducer of the motor, uncoupled the release of the ATP hydrolysis product, inorganic phosphate (Pi), from dependency on actin binding and destroyed the ability of single dimeric molecules to move processively on actin filaments. We observed that processive movement is rescued if ATP is added to the mutant dimer following binding of both heads to actin in the absence of ATP, demonstrating that the mutation selectively destroys the initiation of processive runs at physiological ATP levels. A drug (omecamtiv) that accelerates the actin-activated activity of cardiac myosin was able to rescue processivity of the D179Y mutant dimers at physiological ATP concentrations by slowing the actin-independent release of Pi. Thus, it may be possible to create myosin VI-specific drugs that rescue the function of deafness-causing mutations.


Assuntos
Actinas/metabolismo , Surdez/genética , Mutação/genética , Cadeias Pesadas de Miosina/genética , Difosfato de Adenosina/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Humanos , Cinética , Camundongos , Modelos Biológicos , Modelos Moleculares , Proteínas Mutantes/metabolismo , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/metabolismo , Multimerização Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Sus scrofa , Ureia/análogos & derivados , Ureia/farmacologia
14.
Proc Natl Acad Sci U S A ; 110(51): 20443-8, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24248336

RESUMO

Specific recognition of the cargo that molecular motors transport or tether to cytoskeleton tracks allows them to perform precise cellular functions at particular times and positions in cells. However, very little is known about how evolution has favored conservation of functions for some isoforms, while also allowing for the generation of new recognition sites and specialized cellular functions. Here we present several crystal structures of the myosin Va or the myosin Vb globular tail domain (GTD) that gives insights into how the motor is linked to the recycling membrane compartments via Rab11 or to the melanosome membrane via recognition of the melanophilin adaptor that binds to Rab27a. The structures illustrate how the Rab11-binding site has been conserved during evolution and how divergence at another site of the GTD allows more specific interactions such as the specific recognition of melanophilin by the myosin Va isoform. With atomic structural insights, these structures also show how either the partner or the GTD structural plasticity upon association is critical for selective recruitment of the motor.


Assuntos
Cadeias Pesadas de Miosina/química , Miosina Tipo V/química , Proteínas rab de Ligação ao GTP/química , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cristalografia por Raios X , Humanos , Membranas Intracelulares/química , Membranas Intracelulares/metabolismo , Melanossomas/química , Melanossomas/genética , Melanossomas/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/genética , Miosina Tipo V/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP
15.
Mol Cell ; 48(1): 75-86, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22940248

RESUMO

Myosin VI is the only known reverse-direction myosin motor. It has an unprecedented means of amplifying movements within the motor involving rearrangements of the converter subdomain at the C terminus of the motor and an unusual lever arm projecting from the converter. While the average step size of a myosin VI dimer is 30-36 nm, the step size is highly variable, presenting a challenge to the lever arm mechanism by which all myosins are thought to move. Herein, we present structures of myosin VI that reveal regions of compliance that allow an uncoupling of the lead head when movement is modeled on actin. The location of the compliance restricts the possible actin binding sites and predicts the observed stepping behavior. The model reveals that myosin VI, unlike plus-end directed myosins, does not use a pure lever arm mechanism, but instead steps with a mechanism analogous to the kinesin neck-linker uncoupling model.


Assuntos
Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/metabolismo , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/metabolismo , Actinas/química , Actinas/metabolismo , Animais , Sítios de Ligação , Fenômenos Biofísicos , Calmodulina/química , Calmodulina/metabolismo , Complacência (Medida de Distensibilidade) , Cristalografia por Raios X , Modelos Biológicos , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Miosinas/química , Miosinas/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Suínos
17.
Nat Genet ; 44(4): 456-60, S1-3, 2012 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-22406640

RESUMO

Familial hyperkalemic hypertension (FHHt) is a Mendelian form of arterial hypertension that is partially explained by mutations in WNK1 and WNK4 that lead to increased activity of the Na(+)-Cl(-) cotransporter (NCC) in the distal nephron. Using combined linkage analysis and whole-exome sequencing in two families, we identified KLHL3 as a third gene responsible for FHHt. Direct sequencing of 43 other affected individuals revealed 11 additional missense mutations that were associated with heterogeneous phenotypes and diverse modes of inheritance. Polymorphisms at KLHL3 were not associated with blood pressure. The KLHL3 protein belongs to the BTB-BACK-kelch family of actin-binding proteins that recruit substrates for Cullin3-based ubiquitin ligase complexes. KLHL3 is coexpressed with NCC and downregulates NCC expression at the cell surface. Our study establishes a role for KLHL3 as a new member of the complex signaling pathway regulating ion homeostasis in the distal nephron and indirectly blood pressure.


Assuntos
Proteínas de Transporte/genética , Transporte de Íons/genética , Néfrons/metabolismo , Pseudo-Hipoaldosteronismo/genética , Simportadores de Cloreto de Sódio/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Sequência de Bases , Pressão Sanguínea/genética , Criança , Feminino , Humanos , Rim/metabolismo , Masculino , Proteínas dos Microfilamentos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Pseudo-Hipoaldosteronismo/metabolismo , Pseudo-Hipoaldosteronismo/fisiopatologia , Análise de Sequência de DNA , Transdução de Sinais , Simportadores de Cloreto de Sódio/genética , Adulto Jovem
18.
FEBS J ; 279(4): 551-62, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22171985

RESUMO

Molecular motors such as myosins are allosteric enzymes that power essential motility functions in the cell. Structural biology is an important tool for deciphering how these motors work. Myosins produce force upon the actin-driven conformational changes controlling the sequential release of the hydrolysis products of ATP (Pi followed by ADP). These conformational changes are amplified by a 'lever arm', which includes the region of the motor known as the converter and the adjacent elongated light chain binding region. Analysis of four structural states of the motor provides a detailed understanding of the rearrangements and pathways of communication in the motor that are necessary for detachment from the actin track and repriming of the motor. However, the important part of the cycle in which force is produced remains enigmatic and awaits new high-resolution structures. The value of a structural approach is particularly evident from clues provided by the structural states of the reverse myosin VI motor. Crystallographic structures have revealed that rearrangements within the converter subdomain occur, which explains why this myosin can produce a large stroke in the opposite direction to all other myosins, despite a very short lever arm. By providing a detailed understanding of the motor rearrangements, structural biology will continue to reveal essential information and help solve current enigma, such as how actin promotes force production, how motors are tuned for specific cellular roles or how motor/cargo interactions regulate the function of myosin in the cell.


Assuntos
Fenômenos Fisiológicos Celulares , Proteínas Motores Moleculares/química , Cadeias Pesadas de Miosina/química , Actinas/química , Actinas/metabolismo , Difosfato de Adenosina/química , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/química , Cálcio/metabolismo , Calmodulina/química , Calmodulina/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Proteínas Motores Moleculares/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
19.
J Biol Chem ; 286(13): 11716-23, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21278381

RESUMO

Myosin VI is unique in its directionality among myosin superfamily members and also displays a slow and strain-dependent rate of ATP binding that allows for gating between its heads. In this study we demonstrate that leucine 310 is positioned by a class VI-specific insert, insert-1, so as to account for the selective hindrance of ATP versus ADP binding. Mutation of leucine 310 to glycine removes all influence of insert-1 on ATP binding. Furthermore, by analyzing myosin VI structures with either leucine 310 substituted to a glycine or complete removal of insert-1, we conclude that nucleotides may initially bind to myosin by their purine rings before docking their phosphate moieties. Otherwise, insert-1 could not exert a differential influence on ATP versus ADP binding.


Assuntos
Difosfato de Adenosina/química , Trifosfato de Adenosina/química , Mutação de Sentido Incorreto , Cadeias Pesadas de Miosina/química , Difosfato de Adenosina/genética , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Substituição de Aminoácidos , Animais , Humanos , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...