Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 13: 845407, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36117693

RESUMO

Angiogenesis, the outgrowth of new blood vessels from existing vasculature, is critical during development, tissue formation, and wound healing. In response to vascular endothelial growth factors (VEGFs), endothelial cells are activated to proliferate and move towards the signal, extending the vessel. These events are directed by VEGF-VEGF receptor (Vegfr2) signal transduction, which in turn is modulated by heparan sulfate proteoglycans (HSPGs). HSPGs are glycoproteins covalently attached to HS glycosaminoglycan chains. Transmembrane protein 184a (Tmem184a) has been recently identified as a heparin receptor, which is believed to bind heparan sulfate chains in vivo. Therefore, Tmem184a has the potential to fine-tune interactions between VEGF and HS, modulating Vegfr2-dependent angiogenesis. The function of Tmem184a has been investigated in the regenerating zebrafish caudal fin, but its role has yet to be evaluated during developmental angiogenesis. Here we provide insights into how Tmem184a contributes to the proper formation of the vasculature in zebrafish embryos. First, we find that knockdown of Tmem184a causes a reduction in the number of intact intersegmental vessels (ISVs) in the zebrafish embryo. This phenotype mimics that of vegfr2b knockout mutants, which have previously been shown to exhibit severe defects in ISV development. We then test the importance of HS interactions by removing the binding domain within the Tmem184a protein, which has a negative effect on angiogenesis. Tmem184a is found to act synergistically with Vegfr2b, indicating that the two gene products function in a common pathway to modulate angiogenesis. Moreover, we find that knockdown of Tmem184a leads to an increase in endothelial cell proliferation but a decrease in the amount of VE-cadherin present. Together, these findings suggest that Tmem184a is necessary for ISVs to organize into mature, complete vessels.

2.
Glycobiology ; 31(5): 593-602, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-33021626

RESUMO

Glypican-1 and its heparan sulfate (HS) chains play important roles in modulating many biological processes including growth factor signaling. Glypican-1 is bound to a membrane surface via a glycosylphosphatidylinositol (GPI)-anchor. In this study, we used all-atom molecular modeling and simulation to explore the structure, dynamics, and interactions of GPI-anchored glypican-1, three HS chains, membranes, and ions. The folded glypican-1 core structure is stable, but has substantial degrees of freedom in terms of movement and orientation with respect to the membrane due to the long unstructured C-terminal region linking the core to the GPI-anchor. With unique structural features depending on the extent of sulfation, high flexibility of HS chains can promote multi-site interactions with surrounding molecules near and above the membrane. This study is a first step toward all-atom molecular modeling and simulation of the glycocalyx, as well as its modulation of interactions between growth factors and their receptors.


Assuntos
Membrana Celular/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Glipicanas/metabolismo , Heparitina Sulfato/metabolismo , Termodinâmica , Membrana Celular/química , Biologia Computacional , Glicosilfosfatidilinositóis/química , Glipicanas/química , Heparitina Sulfato/química , Humanos , Modelos Moleculares , Estrutura Molecular
3.
Am J Physiol Cell Physiol ; 318(3): C463-C475, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31891520

RESUMO

Published studies indicate that TMEM184A is a heparin receptor that interacts with and transduces stimulation from heparin in vascular cells. Previous studies have indicated that heparin increases endothelial nitric oxide synthase (eNOS) activity in bovine endothelial cells. However, the precise mechanism remains unknown. In this study, we investigated the impact of heparin treatment and TMEM184A on eNOS's activation and the role of eNOS in heparin signaling in the cloned A7r5 rat vascular smooth muscle cell line and confirmed results in endothelial cells. We employed a combination of TMEM184A knockdown A7r5 cells along with transient eNOS knockdown and enzyme inhibitor strategies. The results indicate that heparin induces phosphorylation of eNOS. eNOS can be immunoprecipitated with TMEM184A and is internalized to the perinuclear region in a TMEM184A-dependent manner in response to heparin. We also examined how heparin treatment leads to phosphorylation of eNOS and confirmed that TMEM184A and Ca2+ were required to mediate heparin-elicited eNOS phosphorylation. Evidence supporting the involvement of transient receptor potential cation channel subfamily V member 4 with TMEM184A in this eNOS activation process is also presented.


Assuntos
Músculo Liso Vascular/citologia , Músculo Liso Vascular/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Anticoagulantes/farmacologia , Bovinos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Heparina/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Ratos , Receptores de Superfície Celular/agonistas
4.
Front Physiol ; 8: 671, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28936181

RESUMO

Transmembrane protein 184A (TMEM184A) was recently identified as the heparin receptor in vascular cells. Heparin binds specifically to TMEM184A and induces anti-proliferative signaling in vitro. Though it is highly conserved, the physiological function of TMEM184A remains unknown. The objective of this study was to investigate the expression and effects on vascular regeneration of TMEM184A using the adult zebrafish regenerating caudal fin as an in vivo model. Here, we show that Tmem184a is expressed in vascular endothelial cells (ECs) of mature and regenerating zebrafish fins. Transient morpholino (MO)-mediated knockdown of Tmem184a using two validated MOs results in tangled regenerating vessels that do not grow outward and limit normal overall fin regeneration. A significant increase in EC proliferation is observed. Consistent with in vitro work with tissue culture vascular cells, heparin has the opposite effect and decreases EC proliferation which also hinders overall fin regeneration. Collectively, our study suggests that Tmem184a is a novel regulator of angiogenesis.

5.
J Vis Exp ; (120)2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28287514

RESUMO

When novel proteins are identified through affinity-based isolation and bioinformatics analysis, they are often largely uncharacterized. Antibodies against specific peptides within the predicted sequence allow some localization experiments. However, other possible interactions with the antibodies often cannot be excluded. This situation provided an opportunity to develop a set of assays dependent on the protein sequence. Specifically, a construct containing the gene sequence coupled to the GFP coding sequence at the C-terminal end of the protein was obtained and employed for these purposes. Experiments to characterize localization, ligand affinity, and gain of function were originally designed and carried out to confirm the identification of TMEM184A as a heparin receptor1. In addition, the construct can be employed for studies addressing membrane topology questions and detailed protein-ligand interactions. The present report presents a range of experimental protocols based on the GFP-TMEM184A construct expressed in vascular cells that could easily be adapted for other novel proteins.


Assuntos
Bioensaio/métodos , Proteínas de Fluorescência Verde/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/fisiologia , Sequência de Aminoácidos , Proteínas de Membrana , Proteínas Recombinantes de Fusão/metabolismo , Análise de Sequência de DNA , Proteínas de Transporte Vesicular
6.
J Biol Chem ; 291(10): 5342-54, 2016 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-26769965

RESUMO

Despite the large number of heparin and heparan sulfate binding proteins, the molecular mechanism(s) by which heparin alters vascular cell physiology is not well understood. Studies with vascular smooth muscle cells (VSMCs) indicate a role for induction of dual specificity phosphatase 1 (DUSP1) that decreases ERK activity and results in decreased cell proliferation, which depends on specific heparin binding. The hypothesis that unfractionated heparin functions to decrease inflammatory signal transduction in endothelial cells (ECs) through heparin-induced expression of DUSP1 was tested. In addition, the expectation that the heparin response includes a decrease in cytokine-induced cytoskeletal changes was examined. Heparin pretreatment of ECs resulted in decreased TNFα-induced JNK and p38 activity and downstream target phosphorylation, as identified through Western blotting and immunofluorescence microscopy. Through knockdown strategies, the importance of heparin-induced DUSP1 expression in these effects was confirmed. Quantitative fluorescence microscopy indicated that heparin treatment of ECs reduced TNFα-induced increases in stress fibers. Monoclonal antibodies that mimic heparin-induced changes in VSMCs were employed to support the hypothesis that heparin was functioning through interactions with a receptor. Knockdown of transmembrane protein 184A (TMEM184A) confirmed its involvement in heparin-induced signaling as seen in VSMCs. Therefore, TMEM184A functions as a heparin receptor and mediates anti-inflammatory responses of ECs involving decreased JNK and p38 activity.


Assuntos
Fosfatase 1 de Especificidade Dupla/metabolismo , Células Endoteliais/metabolismo , Heparina/farmacologia , Receptores de Superfície Celular/metabolismo , Fibras de Estresse/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Bovinos , Linhagem Celular , Fosfatase 1 de Especificidade Dupla/genética , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Humanos , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Ratos , Receptores de Superfície Celular/genética , Proteínas de Transporte Vesicular , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
J Biol Chem ; 291(10): 5326-41, 2016 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-26769966

RESUMO

Vascular cell responses to exogenous heparin have been documented to include decreased vascular smooth muscle cell proliferation following decreased ERK pathway signaling. However, the molecular mechanism(s) by which heparin interacts with cells to induce those responses has remained unclear. Previously characterized monoclonal antibodies that block heparin binding to vascular cells have been found to mimic heparin effects. In this study, those antibodies were employed to isolate a heparin binding protein. MALDI mass spectrometry data provide evidence that the protein isolated is transmembrane protein 184A (TMEM184A). Commercial antibodies against three separate regions of the TMEM184A human protein were used to identify the TMEM184A protein in vascular smooth muscle cells and endothelial cells. A GFP-TMEM184A construct was employed to determine colocalization with heparin after endocytosis. Knockdown of TMEM184A eliminated the physiological responses to heparin, including effects on ERK pathway activity and BrdU incorporation. Isolated GFP-TMEM184A binds heparin, and overexpression results in additional heparin uptake. Together, these data support the identification of TMEM184A as a heparin receptor in vascular cells.


Assuntos
Heparina/farmacologia , Músculo Liso Vascular/metabolismo , Receptores de Superfície Celular/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Humanos , Dados de Sequência Molecular , Músculo Liso Vascular/efeitos dos fármacos , Ligação Proteica , Transporte Proteico , Ratos , Receptores de Superfície Celular/genética , Transdução de Sinais
8.
J Cell Physiol ; 229(12): 2142-52, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24911927

RESUMO

Published data provide strong evidence that heparin treatment of proliferating vascular smooth muscle cells results in decreased signaling through the ERK pathway and decreases in cell proliferation. In addition, these changes have been shown to be mimicked by antibodies that block heparin binding to the cell surface. Here, we provide evidence that the activity of protein kinase G is required for these heparin effects. Specifically, a chemical inhibitor of protein kinase G, Rp-8-pCPT-cGMS, eliminates heparin and anti-heparin receptor antibody effects on bromodeoxyuridine incorporation into growth factor-stimulated cells. In addition, protein kinase G inhibitors decrease heparin effects on ERK activity, phosphorylation of the transcription factor Elk-1, and heparin-induced MKP-1 synthesis. Although transient, the levels of cGMP increase in heparin treated cells. Finally, knock down of protein kinase G also significantly decreases heparin effects in growth factor-activated vascular smooth muscle cells. Together, these data indicate that heparin effects on vascular smooth muscle cell proliferation depend, at least in part, on signaling through protein kinase G.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Heparina/metabolismo , Músculo Liso Vascular/metabolismo , Animais , GMP Cíclico/administração & dosagem , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Heparina/administração & dosagem , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Receptores de Superfície Celular/metabolismo , Tionucleotídeos/administração & dosagem
9.
Mol Reprod Dev ; 80(6): 488-502, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23661501

RESUMO

Sperm CRISP2 has been proposed to be involved in sperm-egg fusion. After the acrosome reaction, it appears at the equatorial segment (EqS) of human sperm; the mechanism underlying the appearance of CRISP2 at the EqS remains unknown, though. Here, we provide evidence showing the re-association of sperm acrosomal CRISP2 at the EqS during the acrosome reaction. Results showed that F-actin is not involved in the relocalization of CRISP2. We found that basic, but not acidic, conditions can solubilize CRISP2 from sperm cells, suggesting that CRISP2 is a component of the acrosome and that it is released from the acrosome during the acrosome reaction. Purified, biotinylated human sperm acrosomal CRISP2 binds to the EqS of acrosome-reacted sperm in a dose-dependent manner, revealing that CRISP2 detected at the EqS of acrosome-reacted sperm comes from the population stored in the acrosome. The association of CRISP2 at the EqS is very strong, and does not depend on ionic interactions or intermolecular disulfide bonds. Interestingly, the restriction of CRISP2 at the EqS was diminished when EGTA was present in the media, indicating that Ca(2+) is required for maintaining CRISP2 at the EqS. This study supports the possibility that CRISP2 may help modify the EqS membrane to make this domain fusion-competent.


Assuntos
Reação Acrossômica/fisiologia , Acrossomo/metabolismo , Glicoproteínas/metabolismo , Acrossomo/química , Acrossomo/efeitos dos fármacos , Reação Acrossômica/efeitos dos fármacos , Actinas/metabolismo , Adulto , Calcimicina/farmacologia , Moléculas de Adesão Celular , Glicoproteínas/análise , Glicoproteínas/química , Humanos , Ionóforos/farmacologia , Masculino , Microscopia de Fluorescência , Estabilidade Proteica , Espermatozoides/química , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo
10.
Mol Reprod Dev ; 80(4): 273-85, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23426913

RESUMO

Sperm-associated α-L-fucosidases have been implicated in fertilization in many species. Previously, we documented the existence of α-L-fucosidase in mouse cauda epididymal contents, and showed that sperm-associated α-L-fucosidase is cryptically stored within the acrosome and reappears within the sperm equatorial segment after the acrosome reaction. The enrichment of sperm membrane-associated α-L-fucosidase within the equatorial segment of acrosome-reacted cells implicates its roles during fertilization. Here, we document the absence of α-L-fucosidase in mouse oocytes and early embryos, and define roles of sperm associated α-L-fucosidase in fertilization using specific inhibitors and competitors. Mouse sperm were pretreated with deoxyfuconojirimycin (DFJ, an inhibitor of α-L-fucosidase) or with anti-fucosidase antibody; alternatively, mouse oocytes were pretreated with purified human liver α-L-fucosidase. Five-millimolar DFJ did not inhibit sperm-zona pellucida (ZP) binding, membrane binding, or fusion and penetration, but anti-fucosidase antibody and purified human liver α-L-fucosidase significantly decreased the frequency of these events. To evaluate sperm-associated α-L-fucosidase enzyme activity in post-fusion events, DFJ-pretreated sperm were microinjected into oocytes, and 2-pronuclear (2-PN) embryos were treated with 5 mM DFJ with no significant effects, suggesting that α-L-fucosidase enzyme activity does not play a role in post-fusion events and/or early embryo development in mice. The recognition and binding of mouse sperm to the ZP and oolemma involves the glycoprotein structure of α-L-fucosidase, but not its catalytic action. These observations suggest that deficits in fucosidase protein and/or the presence of anti-fucosidase antibody may be responsible for some types of infertility.


Assuntos
Reação Acrossômica/fisiologia , Acrossomo/metabolismo , Blastocisto/metabolismo , Oócitos/metabolismo , alfa-L-Fucosidase/metabolismo , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacologia , Reação Acrossômica/efeitos dos fármacos , Animais , Blastocisto/citologia , Feminino , Humanos , Infertilidade/etiologia , Infertilidade/metabolismo , Masculino , Camundongos , Oócitos/citologia , Álcoois Açúcares/farmacologia , alfa-L-Fucosidase/antagonistas & inibidores
11.
J Cell Biochem ; 114(4): 782-95, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23060131

RESUMO

Vascular endothelial cells and their actin microfilaments align in the direction of fluid shear stress (FSS) in vitro and in vivo. To determine whether cofilin, an actin severing protein, is required in this process, the levels of phospho-cofilin (serine-3) were evaluated in cells exposed to FSS. Phospho-cofilin levels decreased in the cytoplasm and increased in the nucleus during FSS exposure. This was accompanied by increased nuclear staining for activated LIMK, a cofilin kinase. Blocking stress kinases JNK and p38, known to play roles in actin realignment during FSS, decreased cofilin phosphorylation under static conditions, and JNK inhibition also resulted in decreased phospho-cofilin during FSS exposure. Inhibition of dynamic changes in cofilin phosphorylation through cofilin mutants decreased correct actin realignment. The mutants also decreased barrier integrity as did inhibition of the stress kinases. These results identify the importance of cofilin in the process of actin alignment and the requirement for actin realignment in endothelial barrier integrity during FSS.


Assuntos
Actinas/metabolismo , Cofilina 1/metabolismo , Células Endoteliais/metabolismo , Estresse Mecânico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antracenos/farmacologia , Antígenos CD/metabolismo , Fenômenos Biomecânicos , Caderinas/metabolismo , Permeabilidade Capilar , Bovinos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Imidazóis/farmacologia , Junções Intercelulares , Quinases Lim/metabolismo , MAP Quinase Quinase 4/antagonistas & inibidores , MAP Quinase Quinase 4/metabolismo , Fosforilação , Piridinas/farmacologia , Serina/metabolismo , beta Catenina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
12.
J Cell Biochem ; 110(2): 382-91, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20235148

RESUMO

The ability of heparin to block proliferation of vascular smooth muscle cells has been well documented. It is clear that heparin treatment can decrease the level of ERK activity in vascular smooth muscle cells that are sensitive to heparin. In this study, the mechanism by which heparin induces decreases in ERK activity was investigated by evaluating the dual specificity phosphatase, MKP-1, in heparin treated cells. Heparin induced MKP-1 synthesis in a time and concentration dependent manner. The time-course of MKP-1 expression correlated with the decrease in ERK activity. Over the same time frame, heparin treatment did not result in decreases in MEK-1 activity which could have, along with constitutive phosphatase activity, accounted for the decrease in ERK activity. Antibodies against a heparin receptor also induced the synthesis of MKP-1 along with decreasing ERK activity. Blocking either phosphatase activity or synthesis also blocked heparin-induced decreases in ERK activity. Consistent with a role for MKP-1, a nuclear phosphatase, heparin treated cells exhibited decreases in nuclear ERK activity more rapidly than cells not treated with heparin. The data support MKP-1 as a heparin-induced phosphatase that dephosphorylates ERK, decreasing ERK activity, in vascular smooth muscle cells.


Assuntos
Fosfatase 1 de Especificidade Dupla/biossíntese , Heparina/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Animais , Anticorpos/imunologia , Western Blotting , Células Cultivadas , Fosfatase 1 de Especificidade Dupla/metabolismo , Ativação Enzimática , Microscopia de Fluorescência , Músculo Liso Vascular/citologia , Músculo Liso Vascular/enzimologia , Receptores de Superfície Celular/imunologia , Suínos
13.
Toxicol Sci ; 111(2): 402-12, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628585

RESUMO

The purpose of this investigation was to determine the utility of fast-twitch skeletal muscle troponin I (fsTnI) and urinary myoglobin (uMB) as biomarkers of skeletal muscle injury in 8-week-old Sprague-Dawley rats. fsTnI and uMB were quantified by enzyme-linked immunosorbent assay and compared with standard clinical assays including creatine kinase, aldolase, aspartate aminotransferase, and histopathological assessments. Detectable levels of uMB were normalized to urinary creatinine to control for differences in renal function. Seven compounds, including those with toxic effects on skeletal muscle, cardiac muscle, or liver, were evaluated. fsTnI was typically nondetectable (< 5.9 ng/ml serum) in vehicle-treated female and male rats but increased in a dose-dependent manner to at least 300 ng/ml in cerivastatin-induced severe fast-twitch specific myotoxicity. Minimal myopathy induced by investigational compounds BMS-600149 and BMS-687453 increased serum fsTnI to about 30-50 ng/ml, suggesting a reasonable dynamic range for detecting mild to severe skeletal muscle toxicity. In direct contrast, fsTnI was only marginally increased relative to population control values in rats treated with triamcinolone acetonide, which produces muscle atrophy or the cardiotoxins isoproterenol and CoCl2. uMB was typically nondetectable (< 1.6 ng/ml urine) in vehicle-treated female and male rats but increased to approximately 140, 300, and 30 ng/mg creatinine in rats treated with cerivastatin, BMS-687453, and triamcinolone acetonide, respectively. Cardiotoxicity also increased uMB in rats treated with isoproterenol and CoCl2 with urine concentrations ranging from 20 to 30 ng/mg creatinine. Severe hepatotoxicity (coumarin) did not significantly affect serum fsTnI or uMB levels. Collectively, these data suggest that fsTnI is specific for skeletal muscle toxicity, whereas uMB is nonspecific, increasing with skeletal muscle and cardiac toxicity. Accordingly, the complement of fsTnI and uMB, in conjunction with standard clinical assays may comprise a useful diagnostic panel for assessing drug-induced myopathy in rats.


Assuntos
Biomarcadores/metabolismo , Músculo Esquelético/efeitos dos fármacos , Mioglobina/metabolismo , Troponina I/metabolismo , Sequência de Aminoácidos , Animais , Feminino , Masculino , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mioglobina/química , Ratos , Ratos Sprague-Dawley , Troponina I/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...