Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Am J Med Genet A ; 188(3): 970-977, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34862840

RESUMO

Nemaline Myopathy (NM) is a disorder of skeletal muscles caused by mutations in sarcomere proteins and characterized by accumulation of microscopic rod or thread-like structures (nemaline bodies) in skeletal muscles. Patients diagnosed with both NM and infantile cardiomyopathy are very rare. A male infant presented, within the first few hours of life, with severe dilated cardiomyopathy, biventricular dysfunction and left ventricular noncompaction. A muscle biopsy on the 8th day of life from the right sternocleidomastoid muscle identified nemaline rods. Whole exome sequencing identified a c.1288 delT (homozygous pathogenic variant) in the CAP2 gene (NM_006366), yielding a CAP2 protein (NP_006357.1) with a p.C430fs. Both parents were heterozygous for the same variant but have no history of heart or muscle disease. Analysis of patient derived fibroblasts and cardiomyocytes derived from induced pluripotent stem cells confirmed the p.C430fs mutation (pathogenic variant), which appears to cause loss of both CAP2 protein and mRNA. The CAP2 gene encodes cyclase associated protein 2, an actin monomer binding and filament depolymerizing protein and CAP2 knockout mice develop severe dilated cardiomyopathy and muscle weakness. The patient underwent a heart transplant at 1 year of age. Heart tissue explanted at that time also showed nemaline rods and additionally disintegration of the myofibrillar structure. Other extra cardiac concerns include mild hypotonia, atrophic and widened scarring. This is the first description of a patient presenting with nemaline myopathy associated with a pathogenic variant of CAP2.


Assuntos
Cardiomiopatia Dilatada , Miopatias da Nemalina , Proteínas Adaptadoras de Transdução de Sinal/genética , Cardiomiopatia Dilatada/complicações , Cardiomiopatia Dilatada/diagnóstico , Cardiomiopatia Dilatada/genética , Homozigoto , Humanos , Recém-Nascido , Masculino , Proteínas de Membrana/genética , Músculo Esquelético/patologia , Mutação , Miopatias da Nemalina/diagnóstico , Miopatias da Nemalina/genética , Miopatias da Nemalina/patologia
2.
Proc Natl Acad Sci U S A ; 116(17): 8397-8402, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30962377

RESUMO

Actin filaments (F-actin) are key components of sarcomeres, the basic contractile units of skeletal muscle myofibrils. A crucial step during myofibril differentiation is the sequential exchange of α-actin isoforms from smooth muscle (α-SMA) and cardiac (α-CAA) to skeletal muscle α-actin (α-SKA) that, in mice, occurs during early postnatal life. This "α-actin switch" requires the coordinated activity of actin regulators because it is vital that sarcomere structure and function are maintained during differentiation. The molecular machinery that controls the α-actin switch, however, remains enigmatic. Cyclase-associated proteins (CAP) are a family of actin regulators with largely unknown physiological functions. We here report a function for CAP2 in regulating the α-actin exchange during myofibril differentiation. This α-actin switch was delayed in systemic CAP2 mutant mice, and myofibrils remained in an undifferentiated stage at the onset of the often excessive voluntary movements in postnatal mice. The delay in the α-actin switch coincided with the onset of motor function deficits and histopathological changes including a high frequency of type IIB ring fibers. Our data suggest that subtle disturbances of postnatal F-actin remodeling are sufficient for predisposing muscle fibers to form ring fibers. Cofilin2, a putative CAP2 interaction partner, has been recently implicated in myofibril actin cytoskeleton differentiation, and the myopathies in cofilin2 and CAP2 mutant mice showed striking similarities. We therefore propose a model in which CAP2 and cofilin2 cooperate in actin regulation during myofibril differentiation.


Assuntos
Citoesqueleto de Actina/fisiologia , Proteínas de Transporte , Diferenciação Celular , Músculo Esquelético , Miofibrilas/fisiologia , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Knockout , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
3.
J Recept Signal Transduct Res ; 40(3): 224-230, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32065019

RESUMO

Context: Reperfusion injury after myocardial infarction is associated with inflammation response and oxidative stress.Aim: The aim of our study is to explore the influence of suppressor of ras val-2 (SRV2) on cardiomyocyte oxidative stress and inflammation response under hypoxia-reoxygenation (HR) injury.Methods: Cell viability was determined via MTT assay. qPCR and western blots were used to analyze the alterations of SRV2 and mammalian STE20-like protein kinases 1 (Mst1). ELISA and qPCR were used to verify the alterations of antioxidants and pro-inflammatory factors.Results: SRV2 was upregulated by HR injury in cardiomyocyte. Interestingly, loss of SRV2 attenuated HR injury-mediated cardiomyocyte damage through inhibiting cardiomyocyte apoptosis. At the molecular levels, SRV2 deletion reduced inflammation and oxidative stress induced by HR injury and thus promoted cardiomyocyte survival. Besides, SRV2 deletion inactivated the Mst1-mROS signaling pathway in cardiomyocyte and thus regulated the inflammation and oxidative stress. Interestingly, overexpression of Mst1 abolished the beneficial effects exerted by SRV2 deletion on HR-mediated cardiomyocyte death.Conclusions: SRV2 upregulation, induced by reperfusion injury, contributes to cardiomyocyte death through the Mst1-mROS signaling pathway.


Assuntos
Apoptose , Inflamação/patologia , Mitocôndrias/metabolismo , Miócitos Cardíacos/patologia , Estresse Oxidativo , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Animais , Hipóxia Celular , Deleção de Genes , Técnicas de Silenciamento de Genes , Miócitos Cardíacos/metabolismo , Oxigênio , Reperfusão , Regulação para Cima
4.
J Cell Sci ; 129(2): 367-79, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26604224

RESUMO

The dynamic assembly and disassembly of actin filaments is essential for the formation and transport of vesicles during endocytosis. In yeast, two types of actin structures, namely cortical patches and cytoplasmic cables, play a direct role in endocytosis, but how their interaction is regulated remains unclear. Here, we show that Srv2/CAP, an evolutionarily conserved actin regulator, is required for efficient endocytosis owing to its role in the formation of the actin patches that aid initial vesicle invagination and of the actin cables that these move along. Deletion of the SRV2 gene resulted in the appearance of aberrant fragmented actin cables that frequently moved past actin patches, the sites of endocytosis. We find that the C-terminal CARP domain of Srv2p is vitally important for the proper assembly of actin patches and cables; we also demonstrate that the N-terminal helical folded domain of Srv2 is required for its localization to actin patches, specifically to the ADP-actin rich region through an interaction with cofilin. These results demonstrate the in vivo roles of Srv2p in the regulation of the actin cytoskeleton during clathrin-mediated endocytosis.


Assuntos
Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas do Citoesqueleto/fisiologia , Endocitose , Proteínas de Saccharomyces cerevisiae/fisiologia , Fatores de Despolimerização de Actina/metabolismo , Estruturas da Membrana Celular/metabolismo , Vesículas Revestidas por Clatrina/metabolismo , Proteínas dos Microfilamentos/metabolismo , Domínios Proteicos , Multimerização Proteica , Transporte Proteico , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/metabolismo
5.
J Biol Chem ; 289(44): 30732-30742, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25228691

RESUMO

Srv2/CAP is a conserved actin-binding protein with important roles in driving cellular actin dynamics in diverse animal, fungal, and plant species. However, there have been conflicting reports about whether the activities of Srv2/CAP are conserved, particularly between yeast and mammalian homologs. Yeast Srv2 has two distinct functions in actin turnover: its hexameric N-terminal-half enhances cofilin-mediated severing of filaments, while its C-terminal-half catalyzes dissociation of cofilin from ADP-actin monomers and stimulates nucleotide exchange. Here, we dissected the structure and function of mouse CAP1 to better understand its mechanistic relationship to yeast Srv2. Although CAP1 has a shorter N-terminal oligomerization sequence compared with Srv2, we find that the N-terminal-half of CAP1 (N-CAP1) forms hexameric structures with six protrusions, similar to N-Srv2. Further, N-CAP1 autonomously binds to F-actin and decorates the sides and ends of filaments, altering F-actin structure and enhancing cofilin-mediated severing. These activities depend on conserved surface residues on the helical-folded domain. Moreover, N-CAP1 enhances yeast cofilin-mediated severing, and conversely, yeast N-Srv2 enhances human cofilin-mediated severing, highlighting the mechanistic conservation between yeast and mammals. Further, we demonstrate that the C-terminal actin-binding ß-sheet domain of CAP1 is sufficient to catalyze nucleotide-exchange of ADP-actin monomers, while in the presence of cofilin this activity additionally requires the WH2 domain. Thus, the structures, activities, and mechanisms of mouse and yeast Srv2/CAP homologs are remarkably well conserved, suggesting that the same activities and mechanisms underlie many of the diverse actin-based functions ascribed to Srv2/CAP homologs in different organisms.


Assuntos
Actinas/química , Proteínas de Transporte/química , Fatores de Despolimerização de Actina/química , Actinas/ultraestrutura , Proteínas Adaptadoras de Transdução de Sinal/química , Difosfato de Adenosina/química , Animais , Proteínas de Transporte/fisiologia , Proteínas do Citoesqueleto/química , Células HEK293 , Humanos , Camundongos , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/química , Especificidade da Espécie
6.
Ann Clin Lab Sci ; 54(3): 335-346, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-39048173

RESUMO

OBJECTIVE: Cardiac dysfunction can result from excessive fibrosis in cardiac fibroblasts (CFs) following an acute myocardial infarction (AMI). SIRT3 has been shown to be associated with numerous cardiovascular diseases. This study aimed to investigate the mechanism by which SIRT3 influences myocardial fibrosis following AMI. METHODS: An AMI model was established in rats and echocardiography was used to assess cardiac systolic function. Triphenyl tetrazolium chloride (TTC) and H&E staining were employed to observe the myocardial histopathological status. Masson trichrome staining was used to detect fibrosis, and the changes in expression of fibrosis-related proteins were detected by Western Blot (WB). In this study, we utilized in vitro cell models stimulated by Ang II to investigate the underlying mechanisms. We employed Transwell and CCK-8 assays to detect the function of CFs. Additionally, we used transmission electron microscopy (TEM) to observe the structural morphology of mitochondria, whereas WB was performed to quantify fibrosis-associated proteins and to assay the changes in SIRT3, SRV2, and Drp1. RESULTS: We observed a significant decrease in the expression of SIRT3 and an increase in mitochondrial fragmentation in rats with AMI. Additionally, we observed upregulation of fibrosis-associated signature proteins and collagen proteins expression. Through the use of vitro Ang II stimulation we observed a downregulation of SIRT3 expression, an increase in mitochondrial fragmentation, and an increase in the proliferation and migration of CFs. Opposite effects were observed when SIRT3 was overexpressed. Additive mitochondrial division agonists were found to stimulate the proliferation and migration of CFs, however, SIRT3 expression was unchanged. Interference with SRV2 and SIRT3 revealed that SIRT3 effectively prevented the expression of SRV2/Drp1, resulting in the inhibition of mitochondrial division and the suppression of CFs proliferative migration. CONCLUSION: In summary, SIRT3 can suppress myocardial fibrosis after acute myocardial infarction by regulating SRV2/Drp1-mediated mitochondrial division.


Assuntos
Fibroblastos , Dinâmica Mitocondrial , Infarto do Miocárdio , Miocárdio , Sirtuína 3 , Animais , Masculino , Ratos , Proliferação de Células , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Dinâmica Mitocondrial/efeitos dos fármacos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/metabolismo , Ratos Sprague-Dawley , Sirtuína 3/metabolismo , Sirtuína 3/genética , Sirtuínas
7.
Curr Biol ; 33(20): 4484-4495.e5, 2023 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-37797614

RESUMO

How actin filaments are spatially organized and remodeled into diverse higher-order networks in vivo is still not well understood. Here, we report an unexpected F-actin "coalescence" activity driven by cyclase-associated protein (CAP) and enhanced by its interactions with actin-binding protein 1 (Abp1). We directly observe S. cerevisiae CAP and Abp1 rapidly transforming branched or linear actin networks by bundling and sliding filaments past each other, maximizing filament overlap, and promoting compaction into bundles. This activity does not require ATP and is conserved, as similar behaviors are observed for the mammalian homologs of CAP and Abp1. Coalescence depends on the CAP oligomerization domain but not the helical folded domain (HFD) that mediates its functions in F-actin severing and depolymerization. Coalescence by CAP-Abp1 further depends on interactions between CAP and Abp1 and interactions between Abp1 and F-actin. Our results are consistent with a mechanism in which the formation of energetically favorable sliding CAP and CAP-Abp1 crosslinks drives F-actin bundle compaction. Roles for CAP and CAP-Abp1 in actin remodeling in vivo are supported by strong phenotypes arising from deletion of the CAP oligomerization domain and by genetic interactions between sac6Δ and an srv2-301 mutant that does not bind Abp1. Together, these observations identify a new actin filament remodeling function for CAP, which is further enhanced by its direct interactions with Abp1.


Assuntos
Actinas , Proteínas de Saccharomyces cerevisiae , Animais , Actinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citoesqueleto de Actina/metabolismo , Mamíferos
8.
Eur J Cell Biol ; 101(2): 151207, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35150966

RESUMO

Cyclase-associated protein (CAP) is an actin binding protein that has been initially described as partner of the adenylyl cyclase in yeast. In all vertebrates and some invertebrate species, two orthologs, named CAP1 and CAP2, have been described. CAP1 and CAP2 are characterized by a similar multidomain structure, but different expression patterns. Several molecular studies clarified the biological function of the different CAP domains, and they shed light onto the mechanisms underlying CAP-dependent regulation of actin treadmilling. However, CAPs are crucial elements not only for the regulation of actin dynamics, but also for signal transduction pathways. During recent years, human genetic studies and the analysis of gene-targeted mice provided important novel insights into the physiological roles of CAPs and their involvement in the pathogenesis of several diseases. In the present review, we summarize and discuss recent progress in our understanding of CAPs' physiological functions, focusing on heart, skeletal muscle and central nervous system as well as their involvement in the mechanisms controlling metabolism. Remarkably, loss of CAPs or impairment of CAPs-dependent pathways can contribute to the pathogenesis of different diseases. Overall, these studies unraveled CAPs complexity highlighting their capability to orchestrate structural and signaling pathways in the cells.


Assuntos
Actinas , Proteínas de Saccharomyces cerevisiae , Proteínas de Capeamento de Actina/metabolismo , Actinas/metabolismo , Animais , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Genética Humana , Humanos , Camundongos , Proteínas dos Microfilamentos/metabolismo , Ligação Proteica , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
9.
Cells ; 10(6)2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204261

RESUMO

Cyclase-associated proteins (CAPs) are evolutionary-conserved actin-binding proteins with crucial functions in regulating actin dynamics, the spatiotemporally controlled assembly and disassembly of actin filaments (F-actin). Mammals possess two family members (CAP1 and CAP2) with different expression patterns. Unlike most other tissues, both CAPs are expressed in the brain and present in hippocampal neurons. We recently reported crucial roles for CAP1 in growth cone function, neuron differentiation, and neuron connectivity in the mouse brain. Instead, CAP2 controls dendritic spine morphology and synaptic plasticity, and its dysregulation contributes to Alzheimer's disease pathology. These findings are in line with a model in which CAP1 controls important aspects during neuron differentiation, while CAP2 is relevant in differentiated neurons. We here report CAP2 expression during neuron differentiation and its enrichment in growth cones. We therefore hypothesized that CAP2 is relevant not only in excitatory synapses, but also in differentiating neurons. However, CAP2 inactivation neither impaired growth cone morphology and motility nor neuron differentiation. Moreover, CAP2 mutant mice did not display any obvious changes in brain anatomy. Hence, differently from CAP1, CAP2 was dispensable for neuron differentiation and brain development. Interestingly, overexpression of CAP2 rescued not only growth cone size in CAP1-deficient neurons, but also their morphology and differentiation. Our data provide evidence for functional redundancy of CAP1 and CAP2 in differentiating neurons, and they suggest compensatory mechanisms in single mutant neurons.


Assuntos
Proteínas de Transporte/metabolismo , Diferenciação Celular/fisiologia , Neurônios/metabolismo , Animais , Camundongos , Neurogênese/fisiologia
10.
Prog Neurobiol ; 202: 102050, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33845164

RESUMO

Neuron connectivity depends on growth cones that navigate axons through the developing brain. Growth cones protrude and retract actin-rich structures to sense guidance cues. These cues control local actin dynamics and steer growth cones towards attractants and away from repellents, thereby directing axon outgrowth. Hence, actin binding proteins (ABPs) moved into the focus as critical regulators of neuron connectivity. We found cyclase-associated protein 1 (CAP1), an ABP with unknown brain function, abundant in growth cones. Super-resolution microscopy and live cell imaging combined with pharmacological approaches on hippocampal neurons from gene-targeted mice revealed a crucial role for CAP1 in actin dynamics that is critical for growth cone morphology and function. Growth cone defects in CAP1 knockout (KO) neurons compromised neuron differentiation and was associated with impaired neuron connectivity in CAP1-KO brains. Mechanistically, by rescue experiments in double KO neurons lacking CAP1 and the key actin regulator cofilin1, we demonstrated that CAP1 was essential for cofilin1 function in growth cone actin dynamics and morphology and vice versa. Together, we identified CAP1 as a novel actin regulator in growth cones that was relevant for neuron connectivity, and we demonstrated functional interdependence of CAP1 and cofilin1 in neuronal actin dynamics and growth cone function.


Assuntos
Actinas , Cones de Crescimento , Animais , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Estado Funcional , Humanos , Camundongos , Neurogênese , Neurônios
11.
Front Cell Dev Biol ; 8: 586631, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072768

RESUMO

Cyclase-associated protein (CAP) has been discovered three decades ago in budding yeast as a protein that associates with the cyclic adenosine monophosphate (cAMP)-producing adenylyl cyclase and that suppresses a hyperactive RAS2 variant. Since that time, CAP has been identified in all eukaryotic species examined and it became evident that the activity in RAS-cAMP signaling is restricted to a limited number of species. Instead, its actin binding activity is conserved among eukaryotes and actin cytoskeleton regulation emerged as its primary function. However, for many years, the molecular functions as well as the developmental and physiological relevance of CAP remained unknown. In the present article, we will compile important recent progress on its molecular functions that identified CAP as a novel key regulator of actin dynamics, i.e., the spatiotemporally controlled assembly and disassembly of actin filaments (F-actin). These studies unraveled a cooperation with ADF/Cofilin and Twinfilin in F-actin disassembly, a nucleotide exchange activity on globular actin monomers (G-actin) that is required for F-actin assembly and an inhibitory function towards the F-actin assembly factor INF2. Moreover, by focusing on selected model organisms, we will review current literature on its developmental and physiological functions, and we will present studies implicating CAP in human pathologies. Together, this review article summarizes and discusses recent achievements in understanding the molecular, developmental and physiological functions of CAP, which led this protein emerge as a novel CAPt'n of actin dynamics.

12.
Aging (Albany NY) ; 12(2): 1417-1432, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31951593

RESUMO

Mitochondrial fission is associated with cardiomyocyte death and myocardial depression, and suppressor of ras val-2 (SRV2) is a newly discovered pro-fission protein. In this study, we examined the mechanisms of SRV2-mediated mitochondrial fission in septic cardiomyopathy. Western blotting, ELISA, and immunofluorescence were used to evaluate mitochondrial function, oxidative balance, energy metabolism and caspase-related death, and siRNA and adenoviruses were used to perform loss- and gain-of-function assays. Our results demonstrated that increased SRV2 expression promotes, while SRV2 knockdown attenuates, cardiomyocyte death in LPS-induced septic cardiomyopathy. Mechanistically, SRV2 activation promoted mitochondrial fission and physiological abnormalities by upregulating oxidative injury, ATP depletion, and caspase-9-related apoptosis. Our results also demonstrated that SRV2 promotes mitochondrial fission via a Mst1-Drp1 axis. SRV2 knockdown decreased Mst1 and Drp1 levels, while Mst1 overexpression abolished the mitochondrial protection and cardiomyocyte survival-promoting effects of SRV2 knockdown. SRV2 is thus a key novel promotor of mitochondrial fission and Mst1-Drp1 axis activity in septic cardiomyopathy.


Assuntos
Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Proteínas Quinases Associadas com Morte Celular/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/genética , Proteínas Proto-Oncogênicas/metabolismo , Animais , Cardiomiopatias/patologia , Sobrevivência Celular/genética , Modelos Animais de Doenças , Lipopolissacarídeos/efeitos adversos , Camundongos , Miócitos Cardíacos/metabolismo
13.
Trop Life Sci Res ; 31(3): 47-61, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33214855

RESUMO

Simian betaretrovirus serotype-2 (SRV-2) is an important pathogenic agent in Asian macaques. It is a potential confounding variable in biomedical research. SRV-2 also provides a valuable viral model compared to other retroviruses which can be used for understanding many aspects of retroviral-host interactions and immunosuppression, infection mechanism, retroviral structure, antiretroviral and vaccine development. In this study, we isolated the gene encoding reverse transcriptase enzyme (RT) of SRV-2 that infected Indonesian cynomolgus monkey (Mf ET1006) and predicted the three dimensional structure model using the iterative threading assembly refinement (I-TASSER) computational programme. This SRV-2 RT Mf ET1006 consisted of 547 amino acids at nucleotide position 3284-4925 of whole genome SRV-2. The polymerase active site located in the finger/palm subdomain characterised by three conserved catalytic aspartates (Asp90, Asp165, Asp166), and has a highly conserved YMDD motif as Tyr163, Met164, Asp165 and Asp166. We estimated that this SRV-2 RT Mf ET1006 structure has the accuracy of template modelling score (TM-score 0.90 ± 0.06) and root mean square deviation (RMSD) 4.7 ± 3.1Å, indicating that this model can be trusted and the accuracy can be seen from the appearance of protein folding in tertiary structure. The superpositionings between SRV-2 RT Mf ET1006 and Human Immunodeficiency Virus-1 (HIV-1) RT were performed to predict the structural in details and to optimise the best fits for illustrations. This SRV-2 RT Mf ET1006 structure model has the highest homology to HIV-1 RT (2B6A.pdb) with estimated accuracy at TM-score 0.911, RMSD 1.85 Å, and coverage of 0.953. This preliminary study of SRV-2 RT Mf ET1006 structure modelling is intriguing and provide some information to explore the molecular characteristic and biochemical mechanism of this enzyme.

14.
Cytoskeleton (Hoboken) ; 72(7): 349-61, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26147656

RESUMO

The patterning of actin cytoskeleton structures in vivo is a product of spatially and temporally regulated polymer assembly balanced by polymer disassembly. While in recent years our understanding of actin assembly mechanisms has grown immensely, our knowledge of actin disassembly machinery and mechanisms has remained comparatively sparse. Saccharomyces cerevisiae is an ideal system to tackle this problem, both because of its amenabilities to genetic manipulation and live-cell imaging and because only a single gene encodes each of the core disassembly factors: cofilin (COF1), Srv2/CAP (SRV2), Aip1 (AIP1), GMF (GMF1/AIM7), coronin (CRN1), and twinfilin (TWF1). Among these six factors, only the functions of cofilin are essential and have been well defined. Here, we investigated the functions of the nonessential actin disassembly factors by performing genetic and live-cell imaging analyses on a combinatorial set of isogenic single, double, triple, and quadruple mutants in S. cerevisiae. Our results show that each disassembly factor makes an important contribution to cell viability, actin organization, and endocytosis. Further, our data reveal new relationships among these factors, providing insights into how they work together to orchestrate actin turnover. Finally, we observe specific combinations of mutations that are lethal, e.g., srv2Δ aip1Δ and srv2Δ crn1Δ twf1Δ, demonstrating that while cofilin is essential, it is not sufficient in vivo, and that combinations of the other disassembly factors perform vital functions.


Assuntos
Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Citoesqueleto de Actina/genética , Fatores de Despolimerização de Actina/genética , Actinas/genética , Actinas/metabolismo , Western Blotting , Técnicas de Inativação de Genes , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
15.
Am J Primatol ; 39(4): 251-261, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-31918505

RESUMO

Natural killer (NK) cell activity was evaluated in three groups of Macaca nemestrina that varied with respect to SAIDS D retrovirus serotype 2 (SRV-2/W) and viremic status. Target cells used were Raji and K562 cells. No significant differences (ANOVA) in mean NK activity were detected among the three groups of animals studied. Using Raji targets, mean LU30/106 ± SEM was 6.3 ± 1.6 for seronegative (V-Ab-) animals, 7.3 ± 1.5 for seropositive (V-Ab+) animals, and 10.2 ± 3.5 for persistently viremic (V + Ab-) animals. Using K562 targets, mean LU30/106 was 7.6 ± 1.7 for seronegative (V-Ab-) animals, 6.5 ± 2.5 for seropositive (V-Ab+) animals, and 5.1 ± 1.9 for persistently viremic (V+Ab-) animals. Percentage blood CD16+ and CD8+cells also were not different in the three groups of animals. NK activity did not always correlate with percentage of CD16+ or CD8+ cells in peripheral blood at the time the assays were done. In persistently viremic animals, there was a strong positive correlation between percent CD16+ and CD8+ cells and NK activity using K562 cells but not Raji cells. Depletion experiments indicated that lysis was mediated by both CD8+ and CD16+ cells with both Raji and K562 cells. However, Raji targets were a better indicator of killing mediated by CD16+ cells. Our studies indicate that M. nemestrina may be classified as high or low responders with regard to NK activity, and there was no correlation with SRV-2/W viral or antibody status. Additionally, our results suggested that group housing of M. nemestrina was usually associated with increased NK activity. In conclusion, studies of NK activity in M. nemestrina should consider target cells used, phenotype of effectors, endogenous (high or low) levels of NK activity in individual animals, and housing conditions. © 1996 Wiley-Liss, Inc.

16.
Cell Adh Migr ; 8(1): 55-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24429384

RESUMO

Cell migration is essential for a variety of fundamental biological processes such as embryonic development, wound healing, and immune response. Aberrant cell migration also underlies pathological conditions such as cancer metastasis, in which morphological transformation promotes spreading of cancer to new sites. Cell migration is driven by actin dynamics, which is the repeated cycling of monomeric actin (G-actin) into and out of filamentous actin (F-actin). CAP (Cyclase-associated protein, also called Srv2) is a conserved actin-regulatory protein, which is implicated in cell motility and the invasiveness of human cancers. It cooperates with another actin regulatory protein, cofilin, to accelerate actin dynamics. Hence, knockdown of CAP1 slows down actin filament turnover, which in most cells leads to reduced cell motility. However, depletion of CAP1 in HeLa cells, while causing reduction in dynamics, actually led to increased cell motility. The increases in motility are likely through activation of cell adhesion signals through an inside-out signaling. The potential to activate adhesion signaling competes with the negative effect of CAP1 depletion on actin dynamics, which would reduce cell migration. In this commentary, we provide a brief overview of the roles of mammalian CAP1 in cell migration, and highlight a likely mechanism underlying the activation of cell adhesion signaling and elevated motility caused by depletion of CAP1.


Assuntos
Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Mamíferos/metabolismo , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA