Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biomed Sci ; 26(1): 12, 2019 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-30678675

RESUMO

BACKGROUND: Osteoclasts (OCs) are motile multinucleated cells derived from differentiation and fusion of hematopoietic progenitors of the monocyte-macrophage lineage that undergo a multistep process called osteoclastogenesis. The biological function of OCs is to resorb bone matrix for controlling bone strength and integrity, which is essential for bone development. The bone resorption function is based on the remodelling of the actin cytoskeleton into an F-actin-rich structure known as the sealing zone for bone anchoring and matrix degradation. Non-muscle caldesmon (l-CaD) is known to participate in the regulation of actin cytoskeletal remodeling, but its function in osteoclastogenesis remains unclear. METHODS/RESULTS: In this study, gain and loss of the l-CaD level in RAW264.7 murine macrophages followed by RANKL induction was used as an experimental approach to examine the involvement of l-CaD in the control of cell fusion into multinucleated OCs in osteoclastogenesis. In comparison with controls, l-CaD overexpression significantly increased TRAP activity, actin ring structure and mineral substrate resorption in RANKL-induced cells. In contrast, gene silencing against l-CaD decreased the potential for RANKL-induced osteoclastogenesis and mineral substrate resorption. In addition, OC precursor cells with l-CaD overexpression and gene silencing followed by RANKL induction caused 13% increase and 24% decrease, respectively, in cell fusion index. To further understand the mechanistic action of l-CaD in the modulation of OC fusion, atomic force microscopy was used to resolve the mechanical changes of cell spreading and adhesion force in RANKL-induced cells with and without l-CaD overexpression or gene silencing. CONCLUSIONS: l-CaD plays a key role in the regulation of actin cytoskeletal remodeling for the formation of actin ring structure at the cell periphery, which may in turn alter the mechanical property of cell-spreading and cell surface adhesion force, thereby facilitating cell-cell fusion into multinucleated OCs during osteoclastogenesis.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Diferenciação Celular , Osteoclastos/metabolismo , Osteogênese , Animais , Macrófagos/metabolismo , Camundongos , Células RAW 264.7
2.
J Muscle Res Cell Motil ; 34(5-6): 441-6, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24264290

RESUMO

Dr. John Gergely passed away on July 26, 2013 after a long and distinguished career. His publications spanned 67 years. He founded the Department of Muscle Research in the Retina Foundation (which later became the Boston Biomedical Research Institute) and served as director for 34 years. Dr. Gergely served on the editorial boards of ten scientific journals. He was elected as a Fellow of both the Biophysical Society and the American Association for the Advancement of Science. Dr. Gergely made major contributions concerning muscle protein structure and function. He was best known for his work on the troponin complex. The insights of John and his associates have provided the foundation for our understanding of calcium regulation in skeletal and cardiac muscle.


Assuntos
Bioquímica/história , Fisiologia/história , Pesquisa Biomédica , História do Século XX , História do Século XXI , Humanos , Proteínas Musculares/química
3.
Biochemistry ; 52(36): 6240-8, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23947723

RESUMO

The motor function of smooth muscle myosin (SmM) is regulated by phosphorylation of the regulatory light chain (RLC) bound to the neck region of the SmM heavy chain. It is generally accepted that unphosphorylated RLC induces interactions between the two heads and between the head and the tail, thus inhibiting the motor activity of SmM, whereas phosphorylation of RLC interrupts those interactions, thus reversing the inhibition and restoring the motor activity to the maximal value. One assumption of this model is that single-headed SmM is fully active regardless of phosphorylation. To re-evaluate this model, we produced a number of SmM constructs with coiled coils of various lengths and examined their structure and regulation. With these constructs we identified the segment in the coiled-coil key for the formation of a stable double-headed structure. In agreement with the current model, we found that the actin-activated ATPase activity of unphosphorylated SmM increased with shortening of the coiled-coil. However, contrary to the current model, we found that the actin-activated ATPase activity of phosphorylated SmM decreased with shortening coiled-coil and only the stable double-headed SmM was fully activated by phosphorylation. These results indicate that single-headed SmM is neither fully active nor fully inhibited. Based on our findings, we propose that cooperation between the two heads is essential, not only for the inhibition of unphosphorylated SmM, but also for the activation of phosphorylated SmM.


Assuntos
Cadeias Pesadas de Miosina/metabolismo , Cadeias Leves de Miosina/metabolismo , Subfragmentos de Miosina/metabolismo , Animais , Galinhas , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Microscopia Eletrônica , Músculo Liso/metabolismo , Cadeias Pesadas de Miosina/química , Cadeias Leves de Miosina/química , Subfragmentos de Miosina/química , Fosforilação
4.
Cancer ; 119(22): 4003-11, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23963810

RESUMO

BACKGROUND: A previous comparative tissue proteomics study by the authors of the current study led to the identification of caldesmon (CaD) as one of the proteins associated with cervical metastasis of oral cavity squamous cell carcinoma (OSCC). In the current investigation, the authors focused on the potential functions of CaD in patients with OSCC. METHODS: CaD expression was examined in tissue samples from 155 patients using immunohistochemical analysis. The expression of CaD variants was determined by Western blot analysis and reverse transcriptase-polymerase chain reaction. In addition, the specific effects of CaD gene overexpression and silence were determined in OSCC cell lines. RESULTS: CaD expression was found to be significantly higher in tumor cells from metastatic lymph nodes compared with primary tumor cells, and was nearly absent in normal oral epithelia. Higher CaD expression was found to be correlated with positive N classification, poor differentiation, perineural invasion, and tumor depth (P = .001, P = .029, P = .001, and P = .031, respectively). In survival analyses, OSCC patients with higher CaD expression were found to have poorer prognosis with regard to disease-specific survival and disease-free survival (P = .003 and P = .014, respectively). Multivariate analyses further indicated that higher CaD expression was an independent predictor of disease-specific survival (P = .043). Serum CaD levels were found to be significantly higher in patients with OSCC, but this finding was not associated with clinicopathological manifestations. Data obtained from in vitro suppression, rescue, and overexpression of CaD in OEC-M1 cells indicated that CaD promotes migration and invasive processes in OSCC cells. CONCLUSIONS: The findings of the current study collectively suggest that the low-molecular-weight CaD expression in OSCC tumors is associated with tumor metastasis and patient survival.


Assuntos
Proteínas de Ligação a Calmodulina/biossíntese , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias Bucais/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas de Ligação a Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Técnicas de Cultura de Células , Movimento Celular/fisiologia , Intervalo Livre de Doença , Feminino , Inativação Gênica , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Imuno-Histoquímica , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Prognóstico , Carcinoma de Células Escamosas de Cabeça e Pescoço , Adulto Jovem
5.
Pflugers Arch ; 465(2): 283-94, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23149489

RESUMO

Smooth muscle caldesmon (h-CaD) is an actin- and myosin-binding protein that reversibly inhibits the actomyosin ATPase activity in vitro. To test the function of h-CaD in vivo, we eliminated its expression in mice. The h-CaD-null animals appeared normal and fertile, although the litter size was smaller. Tissues from the homozygotes lacked h-CaD and exhibited upregulation of the non-muscle isoform, l-CaD, in visceral, but not vascular tonic smooth muscles. While the Ca(2+) sensitivity of force generation of h-CaD-deficient smooth muscle remained largely unchanged, the kinetic behavior during relaxation in arteries was different. Both intact and permeabilized arterial smooth muscle tissues from the knockout animals relaxed more slowly than those of the wild type. Since this difference occurred after myosin dephosphorylation was complete, the kinetic effect most likely resulted from slower detachment of unphosphorylated crossbridges. Detailed analyses revealed that the apparently slower relaxation of h-CaD-null smooth muscle was due to an increase in the amplitude of a slower component of the biphasic tension decay. While the identity of this slower process has not been unequivocally determined, we propose it reflects a thin filament state that elicits fewer re-attached crossbridges. Our finding that h-CaD modulates the rate of smooth muscle relaxation clearly supports a role in the control of vascular tone.


Assuntos
Proteínas de Ligação a Calmodulina/genética , Relaxamento Muscular/genética , Músculo Liso Vascular/metabolismo , Animais , Artérias/metabolismo , Artérias/patologia , Artérias/fisiopatologia , Proteínas de Ligação a Calmodulina/metabolismo , Homozigoto , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miosinas/metabolismo , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Regulação para Cima , Vasodilatação/genética
6.
J Biol Chem ; 287(37): 31447-53, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22791711

RESUMO

Actin is a highly ubiquitous protein in eukaryotic cells that plays a crucial role in cell mechanics and motility. Cell motility is driven by assembling actin as polymerizing actin drives cell protrusions in a process closely involving a host of other actin-binding proteins, notably the actin-related protein 2/3 (Arp2/3) complex, which nucleates actin and forms branched filamentous structures. The Arp2/3 complex preferentially binds specific actin networks at the cell leading edge and forms branched filamentous structures, which drive cell protrusions, but the exact regulatory mechanism behind this process is not well understood. Here we show using in vitro imaging and binding assays that a fragment of the actin-binding protein caldesmon added to polymerizing actin increases the Arp2/3-mediated branching activity, whereas it has no effect on branch formation when binding to aged actin filaments. Because this caldesmon effect is shown to be independent of nucleotide hydrolysis and phosphate release from actin, our results suggest a mechanism by which caldesmon maintains newly polymerized actin in a distinct state that has a higher affinity for the Arp2/3 complex. Our data show that this new state does not affect the level of cooperativity of binding by Arp2/3 complex or its distribution on actin. This presents a novel regulatory mechanism by which caldesmon, and potentially other actin-binding proteins, regulates the interactions of actin with its binding partners.


Assuntos
Citoesqueleto de Actina/química , Complexo 2-3 de Proteínas Relacionadas à Actina/química , Citoesqueleto de Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Animais , Proteínas de Ligação a Calmodulina/química , Proteínas de Ligação a Calmodulina/metabolismo , Bovinos , Humanos , Ligação Proteica
7.
Bioarchitecture ; 1(3): 127-133, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21922043

RESUMO

We have previously reported that actin undergoes a conformational transition (which we named "maturation") during polymerization, and that the actin-binding protein, caldesmon (CaD), when added at an early phase of polymerization, interferes with this process (Huang et al. J Biol Chem 2010; 285:71). The pre-transition filament is characterized by relatively low pyrene-fluorescence intensity when pyrene-labeled actin is used as a reporter of subunit assembly into filaments, whereas the mature filament emits a characteristic enhanced fluorescence. Previously reported co-sedimentation experiments suggest that filament formation is not inhibited by the presence of CaD, despite blocking the transition associated with filament maturation. In this study we visualized structural effects of CaD on the assembly of actin filaments by TIRF and electron microscopy. CaD-free actin forms "rough" filaments with irregular edges and indistinct subunit organization during the initial phase (∼20 min under our conditions) of polymerization as reported previously by others (Steinmetz et al. J Cell Biol 1997; 138:559; Galinska-Rakoczy et al. J Mol Biol 2009; 387:869), which most likely correspond to the pre-transition state preceding the maturation step. Later during the polymerization process "mature" filaments exhibit a smoother F-actin appearance with easily detectible double helically arranged actin subunits. While the inclusion of the actin-binding domain of CaD during actin polymerization does not affect the elongation rate, it is associated with a prolonged pre-transition phase, characterized by a delayed alteration (rough to smooth) of the appearance of filaments, consistent with a later onset of the maturation process.

8.
J Biol Chem ; 285(1): 71-9, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19889635

RESUMO

The actin-binding protein caldesmon (CaD) reversibly inhibits smooth muscle contraction. In non-muscle cells, a shorter CaD isoform co-exists with microfilaments in the stress fibers at the quiescent state, but the phosphorylated CaD is found at the leading edge of migrating cells where dynamic actin filament remodeling occurs. We have studied the effect of a C-terminal fragment of CaD (H32K) on the kinetics of the in vitro actin polymerization by monitoring the fluorescence of pyrene-labeled actin. Addition of H32K or its phosphorylated form either attenuated or accelerated the pyrene emission enhancement, depending on whether it was added at the early or the late phase of actin polymerization. However, the CaD fragment had no effect on the yield of sedimentable actin, nor did it affect the actin ATPase activity. Our findings can be explained by a model in which nascent actin filaments undergo a maturation process that involves at least two intermediate conformational states. If present at early stages of actin polymerization, CaD stabilizes one of the intermediate states and blocks the subsequent filament maturation. Addition of CaD at a later phase accelerates F-actin formation. The fact that CaD is capable of inhibiting actin filament maturation provides a novel function for CaD and suggests an active role in the dynamic reorganization of the actin cytoskeleton.


Assuntos
Actinas/química , Actinas/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Animais , Calmodulina/metabolismo , Proteínas de Ligação a Calmodulina/farmacologia , Simulação por Computador , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fluorescência , Humanos , Cinética , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Pirenos/metabolismo , Coelhos , Software , Fatores de Tempo
9.
J Biomed Sci ; 13(2): 159-72, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16453176

RESUMO

The actin-binding protein caldesmon (CaD) exists both in smooth muscle (the heavy isoform, h-CaD) and non-muscle cells (the light isoform, l-CaD). In smooth muscles h-CaD binds to myosin and actin simultaneously and modulates the actomyosin interaction. In non-muscle cells l-CaD binds to actin and stabilizes the actin stress fibers; it may also mediate the interaction between actin and non-muscle myosins. Both h- and l-CaD are phosphorylated in vivo upon stimulation. The major phosphorylation sites of h-CaD when activated by phorbol ester are the Erk-specific sites, modification of which is attenuated by the MEK inhibitor PD98059. The same sites in l-CaD are also phosphorylated when cells are stimulated to migrate, whereas in dividing cells l-CaD is phosphorylated more extensively, presumably by cdc2 kinase. Both Erk and cdc2 are members of the MAPK family. Thus it appears that CaD is a downstream effector of the Ras signaling pathways. Significantly, the phosphorylatable serine residues shared by both CaD isoforms are in the C-terminal region that also contains the actin-binding sites. Biochemical and structural studies indicated that phosphorylation of CaD at the Erk sites is accompanied by a conformational change that partially dissociates CaD from actin. Such a structural change in h-CaD exposes the myosin-binding sites on the actin surface and allows actomyosin interactions in smooth muscles. In the case of non-muscle cells, the change in l-CaD weakens the stability of the actin filament and facilitates its disassembly. Indeed, the level of l-CaD modification correlates very well in a reciprocal manner with the level of actin stress fibers. Since both cell migration and cell division require dynamic remodeling of actin cytoskeleton that leads to cell shape changes, phosphorylation of CaD may therefore serve as a plausible means to regulate these processes. Thus CaD not only links the smooth muscle contractility and non-muscle motility, but also provides a common mechanism for the regulation of cell migration and cell proliferation.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Contração Muscular , Animais , Movimento Celular , Proliferação de Células , Citoesqueleto/metabolismo , Humanos , Músculo Liso/fisiologia , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...