Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Innovation (Camb) ; 5(3): 100626, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38699777

RESUMO

Pancreatic adenocarcinoma (PDAC) is one of the most deadly cancers, characterized by extremely limited therapeutic options and a poor prognosis, as it is often diagnosed during late disease stages. Innovative and selective treatments are urgently needed, since current therapies have limited efficacy and significant side effects. Through proteomics analysis of extracellular vesicles, we discovered an imbalanced distribution of amino acids secreted by PDAC tumor cells. Our findings revealed that PDAC cells preferentially excrete proteins with certain preferential amino acids, including isoleucine and histidine, via extracellular vesicles. These amino acids are associated with disease progression and can be targeted to elicit selective toxicity to PDAC tumor cells. Both in vitro and in vivo experiments demonstrated that supplementation with these specific amino acids effectively eradicated PDAC cells. Mechanistically, we also identified XRN1 as a potential target for these amino acids. The high selectivity of this treatment method allows for specific targeting of tumor metabolism with very low toxicity to normal tissues. Furthermore, we found this treatment approach is easy-to-administer and with sustained tumor-killing effects. Together, our findings reveal that exocytosed amino acids may serve as therapeutic targets for designing treatments of intractable PDAC and potentially offer alternative treatments for other types of cancers.

2.
Eur J Cell Biol ; 102(2): 151316, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37084657

RESUMO

The expression of the angiotensin-converting enzyme 2 (ACE2) is altered in multiple chronic kidney diseases like hypertension and renal fibrosis, where the signaling from the basal membrane proteins is critical for the development and progression of the various pathologies. Integrins are heterodimeric cell surface receptors that have important roles in the progression of these chronic kidney diseases by altering various cell signaling pathways in response to changes in the basement membrane proteins. It is unclear whether integrin or integrin-mediated signaling affects the ACE2 expression in the kidney. The current study tests the hypothesis that integrin ß1 regulates the expression of ACE2 in kidney epithelial cells. The role of integrin ß1 in ACE2 expression in renal epithelial cells was investigated by shRNA-mediated knockdown and pharmacological inhibition. In vivo studies were carried out using epithelial cell-specific deletion of integrin ß1 in the kidneys. Deletion of integrin ß1 from the mouse renal epithelial cells reduced the expression of ACE2 in the kidney. Furthermore, the downregulation of integrin ß1 using shRNA decreased ACE2 expression in human renal epithelial cells. ACE2 expression levels were also decreased in renal epithelial cells and cancer cells when treated with an integrin α2ß1 antagonist, BTT 3033. SARS-CoV-2 viral entry to human renal epithelial cells and cancer cells was also inhibited by BTT 3033. This study demonstrates that integrin ß1 positively regulates the expression of ACE2, which is required for the entry of SARS-CoV-2 into kidney cells.


Assuntos
COVID-19 , Insuficiência Renal Crônica , Humanos , Animais , Camundongos , Integrina beta1/genética , Integrina beta1/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , SARS-CoV-2/metabolismo , COVID-19/metabolismo , COVID-19/patologia , Rim/metabolismo , Rim/patologia , Células Epiteliais/metabolismo , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia
3.
Kidney Int ; 103(1): 144-155, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36273656

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) involves the development and persistent growth of fluid filled kidney cysts. In a recent study, we showed that ADPKD kidney cyst epithelial cells can stimulate the proliferation and differentiation of peri-cystic myofibroblasts. Although dense myofibroblast populations are often found surrounding kidney cysts, their role in cyst enlargement or fibrosis in ADPKD is unclear. To clarify this, we examined the effect of myofibroblast depletion in the Pkd1RC/RC (RC/RC) mouse model of ADPKD. RC/RC;αSMAtk mice that use the ganciclovir-thymidine kinase system to selectively deplete α-smooth muscle actin expressing myofibroblasts were generated. Ganciclovir treatment for four weeks depleted myofibroblasts, reduced kidney fibrosis and preserved kidney function in these mice. Importantly, myofibroblast depletion significantly reduced cyst growth and cyst epithelial cell proliferation in RC/RC;αSMAtk mouse kidneys. Similar ganciclovir treatment did not alter cyst growth or fibrosis in wild-type or RC/RC littermates. In vitro, co-culture with myofibroblasts from the kidneys of patients with ADPKD increased 3D microcyst growth of human ADPKD cyst epithelial cells. Treatment with conditioned culture media from ADPKD kidney myofibroblasts increased microcyst growth and cell proliferation of ADPKD cyst epithelial cells. Further examination of ADPKD myofibroblast conditioned media showed high levels of protease inhibitors including PAI1, TIMP1 and 2, NGAL and TFPI-2, and treatment with recombinant PAI1 and TIMP1 increased ADPKD cyst epithelial cell proliferation in vitro. Thus, our findings show that myofibroblasts directly promote cyst epithelial cell proliferation, cyst growth and fibrosis in ADPKD kidneys, and their targeting could be a novel therapeutic strategy to treat PKD.


Assuntos
Cistos , Rim Policístico Autossômico Dominante , Humanos , Camundongos , Animais , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Miofibroblastos , Células Cultivadas , Rim/patologia , Proliferação de Células , Fibrose , Cistos/tratamento farmacológico , Cistos/patologia , Células Epiteliais/patologia
4.
Int J Mol Sci ; 23(14)2022 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-35886951

RESUMO

Vasopressin type-2 receptor (V2R) is ectopically expressed and plays a pathogenic role in clear cell renal cell carcinoma (ccRCC) tumor cells. Here we examined how V2R signaling within human ccRCC tumor cells (Caki1 cells) stimulates stromal cancer-associated fibroblasts (CAFs). We found that cell culture conditioned media from Caki1 cells increased activation, migration, and proliferation of fibroblasts in vitro, which was inhibited by V2R gene silencing in Caki1 cells. Analysis of the conditioned media and mRNA of the V2R gene silenced and control Caki1 cells showed that V2R regulates the production of CAF-activating factors. Some of these factors were also found to be regulated by YAP in these Caki1 cells. YAP expression colocalized and correlated with V2R expression in ccRCC tumor tissue. V2R gene silencing or V2R antagonist significantly reduced YAP in Caki1 cells. Moreover, the V2R antagonist reduced YAP expression and myofibroblasts in mouse xenograft tumors. These results suggest that V2R plays an important role in secreting pro-fibrotic factors that stimulate fibroblast activation by a YAP-dependent mechanism in ccRCC tumors. Our results demonstrate a novel role for the V2R-YAP axis in the regulation of myofibroblasts in ccRCC and a potential therapeutic target.


Assuntos
Fibroblastos Associados a Câncer , Carcinoma de Células Renais , Neoplasias Renais , Receptores de Vasopressinas , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Fibroblastos Associados a Câncer/efeitos dos fármacos , Fibroblastos Associados a Câncer/metabolismo , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Meios de Cultivo Condicionados , Fibroblastos/metabolismo , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/fisiologia , Vasopressinas/genética , Vasopressinas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
ACS Sens ; 6(12): 4489-4498, 2021 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-34846848

RESUMO

Tumor-derived extracellular vesicles (EVs) are under intensive study for their potential as noninvasive diagnosis biomarkers. Most EV-based cancer diagnostic assays trace supernumerary of a single cancer-associated marker or marker signatures. These types of biomarker assays are either subtype-specific or vulnerable to be masked by high background signals. In this study, we introduce using the ß-sheet richness (BR) of the tumor-derived EVs as an effective way to discriminate EVs originating from malignant and nonmalignant cells, where EV contents are evaluated as a collective attribute rather than single factors. Circular dichroism, Fourier transform infrared spectroscopy, fluorescence staining assays, and a de novo workflow combining proteomics, bioinformatics, and protein folding simulations were employed to validate the collective attribute at both cellular and EV levels. Based on the BR of the tumorous EVs, we integrated immunoprecipitation and fluorescence labeling targeting the circulating tumor-derived EVs in serum and developed the process into a clinical assay, named EvIPThT. The assay can distinguish patients with and without malignant disease in a pilot cohort, with weak correlations to prognosis biomarkers, suggesting the potential for a cancer screening panel with existing prognostic biomarkers to improve overall performance.


Assuntos
Vesículas Extracelulares , Neoplasias Pancreáticas , Biomarcadores Tumorais , Detecção Precoce de Câncer , Humanos , Neoplasias Pancreáticas/diagnóstico , Conformação Proteica em Folha beta
6.
Cell Death Dis ; 12(10): 947, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34650051

RESUMO

Autosomal-dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease and is characterized by progressive growth of fluid-filled cysts. Growth factors binding to receptor tyrosine kinases (RTKs) stimulate cell proliferation and cyst growth in PKD. Nintedanib, a triple RTK inhibitor, targets the vascular endothelial growth-factor receptor (VEGFR), platelet-derived growth-factor receptor (PDGFR), and fibroblast growth-factor receptor (FGFR), and is an approved drug for the treatment of non-small-cell lung carcinoma and idiopathic lung fibrosis. To determine if RTK inhibition using nintedanib can slow ADPKD progression, we tested its effect on human ADPKD renal cyst epithelial cells and myofibroblasts in vitro, and on Pkd1f/fPkhd1Cre and Pkd1RC/RC, orthologous mouse models of ADPKD. Nintedanib significantly inhibited cell proliferation and in vitro cyst growth of human ADPKD renal cyst epithelial cells, and cell viability and migration of human ADPKD renal myofibroblasts. Consistently, nintedanib treatment significantly reduced kidney-to-body-weight ratio, renal cystic index, cystic epithelial cell proliferation, and blood-urea nitrogen levels in both the Pkd1f/fPkhd1Cre and Pkd1RC/RC mice. There was a corresponding reduction in ERK, AKT, STAT3, and mTOR activity and expression of proproliferative factors, including Yes-associated protein (YAP), c-Myc, and Cyclin D1. Nintedanib treatment significantly reduced fibrosis in Pkd1RC/RC mice, but did not affect renal fibrosis in Pkd1f/fPkhd1Cre mice. Overall, these results suggest that nintedanib may be repurposed to effectively slow cyst growth in ADPKD.


Assuntos
Indóis/uso terapêutico , Rim Policístico Autossômico Dominante/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Indóis/farmacologia , Rim/efeitos dos fármacos , Rim/patologia , Camundongos Knockout , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/patologia , Inibidores de Proteínas Quinases/farmacologia , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34638957

RESUMO

The tumor microenvironment plays a critical role in defining the growth and malignancy of solid tumors. Extracellular matrix (ECM) proteins such as collagen, vitronectin, and fibronectin are major components of the tumor microenvironment. Tumor growth-promoting reciprocal interaction between ECM and cytoplasmic proteins is regulated by the cell surface receptors called integrins. This study investigated the mechanism by which integrin ß1 promotes pancreatic tumor growth. In MIA PaCa-2 pancreatic cancer cell line, the loss of integrin ß1 protein reduced the ability of cells to proliferate in a 3D matrix and compromised the ability to form a focal adhesion complex. Decreased expression of integrin α5 was observed in KO cells, which resulted in impaired cell spreading and adhesion on vitronectin and fibronectin. Reduced expression of the integrin-associated protein, kindlin-2 was also recorded. The downregulation of kindlin-2 decreased the phosphorylation of Smad2/3 by reducing the expression of TGF-ß receptor 2. These results unravel a new mechanism of integrin ß1 in tumor growth by modifying the expression of kindlin-2 and TGF-ß receptor 2 signaling.


Assuntos
Carcinogênese/metabolismo , Proliferação de Células/genética , Integrina beta1/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Transdução de Sinais/genética , Carcinogênese/genética , Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Integrina beta1/genética , Neoplasias Pancreáticas/patologia , Transfecção
8.
FASEB J ; 34(6): 7885-7904, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32297675

RESUMO

Nearly 6 million Americans suffer from heart failure. Increased fibrosis contributes to functional decline of the heart that leads to heart failure. Previously, we identified a mechanosensitive protein, small proline-rich repeat 3 (SPRR3), in vascular smooth muscle cells of atheromas. In this study, we demonstrate SPRR3 expression in cardiac fibroblasts which is induced in activated fibroblasts following pressure-induced heart failure. Sprr3 deletion in mice showed preserved cardiac function and reduced interstitial fibrosis in vivo and reduced fibroblast proliferation and collagen expression in vitro. SPRR3 loss resulted in reduced activation of Akt, FAK, ERK, and p38 signaling pathways, which are coordinately regulated by integrins and growth factors. SPRR3 deletion did not impede integrin-associated functions including cell adhesion, migration, or contraction. SPRR3 loss resulted in reduced activation of PDGFRß in fibroblasts. This was not due to the reduced PDGFRß expression levels or decreased binding of the PDGF ligand to PDGFRß. SPRR3 facilitated the association of integrin ß1 with PDGFRß and subsequently fibroblast proliferation, suggesting a role in PDGFRß-Integrin synergy. We postulate that SPRR3 may function as a conduit for the coordinated activation of PDGFRß by integrin ß1, leading to augmentation of fibroblast proliferation and matrix synthesis downstream of biomechanical and growth factor signals.


Assuntos
Proliferação de Células/fisiologia , Proteínas Ricas em Prolina do Estrato Córneo/metabolismo , Fibroblastos/metabolismo , Coração/fisiologia , Integrina beta1/metabolismo , Miocárdio/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Adesão Celular/fisiologia , Colágeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais/fisiologia
9.
J Conserv Dent ; 20(4): 245-250, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29259361

RESUMO

AIM: This study was undertaken to evaluate the efficacy of smear layer removal and nanostructural and chemical changes caused by chitosan and ethylenediaminetetraacetic acid (EDTA) on tooth surface using atomic force microscopic analysis and energy-dispersive X-ray (EDX) analysis. METHODOLOGY: Forty single-rooted premolars were decoronated to a standard length of 15 mm and enlarged to Protaper F3 with irrigation of 1 mL 1% NaOCl and deionized water. Specimens were then divided into 4 groups with 10 samples each and subjected to final rinse with 17% EDTA solution, 0.2% and 0.5% chitosan solution for 1 min. Samples were sectioned into 2 halves. One half of sample from each group were subjected to EDX analysis to check the calcium/phosphate (Ca/P) ratio. The second half of sample from each group subjected to atomic force microscopy (AFM) analysis to study the smear layer removal and nanostructural changes. Statistical analysis was done using ANOVA and Chi-square test. RESULTS: The AFM images showed no difference in the elimination of smear layer. The quantitative analysis using AFM showed EDTA group had significantly higher surface alteration than chitosan. EDX analysis showed that the Ca/P ratio of root dentine in EDTA group is significantly lower than chitosan group. CONCLUSION: Chitosan is an effective chelating agent with less alteration in radicular dentine.

10.
Development ; 144(22): 4148-4158, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28993400

RESUMO

Kidney collecting system development requires integrin-dependent cell-extracellular matrix interactions. Integrins are heterodimeric transmembrane receptors consisting of α and ß subunits; crucial integrins in the kidney collecting system express the ß1 subunit. The ß1 cytoplasmic tail has two NPxY motifs that mediate functions by binding to cytoplasmic signaling and scaffolding molecules. Talins, scaffolding proteins that bind to the membrane proximal NPxY motif, are proposed to activate integrins and to link them to the actin cytoskeleton. We have defined the role of talin binding to the ß1 proximal NPxY motif in the developing kidney collecting system in mice that selectively express a Y-to-A mutation in this motif. The mice developed a hypoplastic dysplastic collecting system. Collecting duct cells expressing this mutation had moderate abnormalities in cell adhesion, migration, proliferation and growth factor-dependent signaling. In contrast, mice lacking talins in the developing ureteric bud developed kidney agenesis and collecting duct cells had severe cytoskeletal, adhesion and polarity defects. Thus, talins are essential for kidney collecting duct development through mechanisms that extend beyond those requiring binding to the ß1 integrin subunit NPxY motif.


Assuntos
Integrina beta1/metabolismo , Morfogênese , Talina/metabolismo , Ureter/citologia , Ureter/embriologia , Junções Aderentes/metabolismo , Motivos de Aminoácidos , Animais , Sítios de Ligação , Adesão Celular , Membrana Celular/metabolismo , Polaridade Celular , Regulação da Expressão Gênica no Desenvolvimento , Integrina beta1/química , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/embriologia , Camundongos Endogâmicos C57BL , Mutação/genética , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Ureter/metabolismo
11.
Elife ; 52016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27929375

RESUMO

Integrins are transmembrane receptors composed of α and ß subunits. Although most integrins contain ß1, canonical activation mechanisms are based on studies of the platelet integrin, αIIbß3. Its inactive conformation is characterized by the association of the αIIb transmembrane and cytosolic domain (TM/CT) with a tilted ß3 TM/CT that leads to activation when disrupted. We show significant structural differences between ß1 and ß3 TM/CT in bicelles. Moreover, the 'snorkeling' lysine at the TM/CT interface of ß subunits, previously proposed to regulate αIIbß3 activation by ion pairing with nearby lipids, plays opposite roles in ß1 and ß3 integrin function and in neither case is responsible for TM tilt. A range of affinities from almost no interaction to the relatively high avidity that characterizes αIIbß3 is seen between various α subunits and ß1 TM/CTs. The αIIbß3-based canonical model for the roles of the TM/CT in integrin activation and function clearly does not extend to all mammalian integrins.


Assuntos
Células Epiteliais/fisiologia , Integrina alfa1/metabolismo , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Multimerização Proteica , Substituição de Aminoácidos , Adesão Celular , Células Cultivadas , Células Epiteliais/química , Humanos , Integrina alfa1/química , Integrina beta1/química , Integrina beta1/genética , Integrina beta3/química , Integrina beta3/genética , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/química , Ligação Proteica
12.
J Biol Chem ; 289(12): 8532-44, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24509849

RESUMO

Epithelial cells lining the gastrointestinal tract and kidney have different abilities to facilitate paracellular and transcellular transport of water and solutes. In the kidney, the proximal tubule allows both transcellular and paracellular transport, while the collecting duct primarily facilitates transcellular transport. The claudins and E-cadherin are major structural and functional components regulating paracellular transport. In this study we present the novel finding that the transmembrane matrix receptors, integrins, play a role in regulating paracellular transport of renal proximal tubule cells. Deleting the integrin ß1 subunit in these cells converts them from a "loose" epithelium, characterized by low expression of E-cadherin and claudin-7 and high expression of claudin-2, to a "tight" epithelium with increased E-cadherin and claudin-7 expression and decreased claudin-2 expression. This effect is mediated by the integrin ß1 cytoplasmic tail and does not entail ß1 heterodimerization with an α-subunit or its localization to the cell surface. In addition, we demonstrate that deleting the ß1 subunit in the proximal tubule of the kidney results in a major urine-concentrating defect. Thus, the integrin ß1 tail plays a key role in regulating the composition and function of tight and adherens junctions that define paracellular transport properties of terminally differentiated renal proximal tubule epithelial cells.


Assuntos
Deleção de Genes , Integrina beta1/genética , Integrina beta1/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Permeabilidade da Membrana Celular , Células Cultivadas , Claudina-2/genética , Claudina-2/metabolismo , Regulação para Baixo , Células Epiteliais/metabolismo , Integrina beta1/análise , Camundongos , Permeabilidade , Regulação para Cima , Urina/química
13.
Biochemistry ; 52(8): 1303-20, 2013 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-23368985

RESUMO

From roughly 1985 through the start of the new millennium, the cutting edge of solution protein nuclear magnetic resonance (NMR) spectroscopy was to a significant extent driven by the aspiration to determine structures. Here we survey recent advances in protein NMR that herald a renaissance in which a number of its most important applications reflect the broad problem-solving capability displayed by this method during its classical era during the 1970s and early 1980s.


Assuntos
Ressonância Magnética Nuclear Biomolecular/métodos , Proteínas/química , Animais , Descoberta de Drogas , História do Século XX , História do Século XXI , Humanos , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular/história , Conformação Proteica , Mapeamento de Interação de Proteínas/história , Mapeamento de Interação de Proteínas/métodos , Proteínas/metabolismo
14.
Mol Cell Biol ; 32(20): 4080-91, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22869523

RESUMO

Loss of ß1 integrin expression inhibits renal collecting-system development. Two highly conserved NPXY motifs in the distal ß1 tail regulate integrin function by associating with phosphtyrosine binding (PTB) proteins, such as talin and kindlin. Here, we define the roles of these two tyrosines in collecting-system development and delineate the structural determinants of the distal ß1 tail using nuclear magnetic resonance (NMR). Mice carrying alanine mutations have moderate renal collecting-system developmental abnormalities relative to ß1-null mice. Phenylalanine mutations did not affect renal collecting-system development but increased susceptibility to renal injury. NMR spectra in bicelles showed the distal ß1 tail is disordered and does not interact with the model membrane surface. Alanine or phenylalanine mutations did not alter ß1 structure or interactions between α and ß1 subunit transmembrane/cytoplasmic domains; however, they did decrease talin and kindlin binding. Thus, these studies highlight the fact that the functional roles of the NPXY motifs are organ dependent. Moreover, the ß1 cytoplasmic tail, in the context of the adjacent transmembrane domain in bicelles, is significantly different from the more ordered, membrane-associated ß3 integrin tail. Finally, tyrosine mutations of ß1 NPXY motifs induce phenotypes by disrupting their interactions with critical integrin binding proteins like talins and kindlins.


Assuntos
Integrina beta1/metabolismo , Túbulos Renais Coletores/crescimento & desenvolvimento , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Linhagem Celular , Citosol/metabolismo , Humanos , Integrina beta1/genética , Integrina beta3/química , Integrina beta3/metabolismo , Proteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Mutação , Proteínas de Neoplasias/química , Ligação Proteica , Conformação Proteica , Talina/química , Tirosina/química , Tirosina/genética , Tirosina/metabolismo
15.
Mol Pharm ; 9(4): 752-61, 2012 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-22221179

RESUMO

Bilayered detergent-lipid assemblies known as bicelles have been widely used as model membranes in structural biological studies and are being explored for wider applications, including pharmaceutical use. Most studies to date have involved the use of concentrated bicelle mixtures, such that little is known about the capacity of bicellar mixtures to be diluted without unwanted transitions to nonisotropic phases. Here, different detergent/lipid mixtures have been explored, leading to the identification of two different families of bicelles for which it is possible to lower the total amphiphile (detergent + lipid) concentration to <1% (w/v) while retaining isotropic assemblies. These include a novel family of bicelles based on mixtures of 6-cyclohexyl-1-hexylphosphocholine (Cyclofos-6) and the lipid dimyristoylphosphatidylcholine (DMPC). Bicelles formed by these mixtures can be diluted to <0.5% and also have attractive biochemical properties. However, a caveat of our results is that the diffusion coefficients measured for the lipid component of the different bicelles tested were seen to be dependent on sample history, even though all samples were optically transparent. This suggests that the phase behavior of bicelles at low lipid-to-detergent ratios may be more complex than previously appreciated.


Assuntos
Detergentes/química , Bicamadas Lipídicas/química , Espectroscopia de Ressonância Magnética , Proteínas de Membrana/química , Micelas
16.
Pediatr Nephrol ; 27(6): 891-900, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21603909

RESUMO

The kidney develops from direct interactions between the ureteric bud and the metanephric mesenchyme. The ureteric bud gives rise to the collecting system and the metanephric mesenchyme to the nephrons. The complex process of renal development which occurs between these embryologically distinct structures is mediated by numerous factors, including the communication of cells with their surrounding extracellular matrix. Integrins are the principal cellular receptors for extracellular matrix proteins, and they play a role in organ and tissue development. In this review we focus on how integrins regulate renal development.


Assuntos
Integrinas/metabolismo , Rim/metabolismo , Transdução de Sinais , Animais , Matriz Extracelular/metabolismo , Humanos , Rim/embriologia , Rim/crescimento & desenvolvimento , Morfogênese
17.
J Biol Chem ; 285(51): 40114-24, 2010 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-20940300

RESUMO

Integrin α1ß1 is a collagen receptor that down-regulates collagen and reactive oxygen species (ROS) production, and mice lacking this receptor show increased ROS levels and exacerbated glomerular sclerosis following injury. Caveolin-1 (Cav-1) is a multifunctional protein that is tyrosine-phosphorylated in response to injury and has been implicated in ROS-mediated injury. Cav-1 interacts with integrins, and integrin α1ß1 binds/activates T cell protein-tyrosine phosphatase (TCPTP), which is homologous to the tyrosine phosphatase PTP1B known to dephosphorylate Cav-1. In this study, we analyzed whether phosphorylated Cav-1 (pCav-1) is a substrate of TCPTP and if integrin α1ß1 is essential for promoting TCPTP-mediated Cav-1 dephosphorylation. We found that Cav-1 phosphorylation is significantly higher in cells lacking integrin α1ß1 at base line and following oxidative stress. Overexpression of TCPTP leads to reduced pCav-1 levels only in cells expressing integrin α1ß1. Using solid phase binding assays, we demonstrated that 1) purified Cav-1 directly interacts with TCPTP and the integrin α1 subunit, 2) pCav-1 is a substrate of TCPTP, and 3) TCPTP-mediated Cav-1 dephosphorylation is highly increased by the addition of purified integrin α1ß1 or an integrin α1 cytoplasmic peptide to which TCPTP has been shown to bind. Thus, our results demonstrate that pCav-1 is a new substrate of TCPTP and that integrin α1ß1 acts as a negative regulator of Cav-1 phosphorylation by activating TCPTP. This could explain the protective function of integrin α1ß1 in oxidative stress-mediated damage and why integrin α1-null mice are more susceptible to fibrosis following injury.


Assuntos
Caveolina 1/metabolismo , Integrina alfa1beta1/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , Animais , Células CHO , Caveolina 1/genética , Colágeno/genética , Colágeno/metabolismo , Cricetinae , Cricetulus , Ativação Enzimática/genética , Fibrose/genética , Células HEK293 , Humanos , Integrina alfa1beta1/genética , Camundongos , Camundongos Mutantes , Estresse Oxidativo/genética , Fosforilação/genética , Proteína Tirosina Fosfatase não Receptora Tipo 2/genética , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
J Biol Chem ; 284(51): 35278-82, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19808673

RESUMO

We have previously reported that the epithelial cell-specific actin-binding protein villin directly associates with phosphatidylinositol 4,5-bisphosphate (PIP(2)) through three binding sites that overlap with actin-binding sites in villin. As a result, association of villin with PIP(2) inhibits actin depolymerization and enhances actin cross-linking by villin. In this study, we demonstrate that these three PIP(2)-binding sites also bind the more hydrophilic phospholipid, lysophosphatidic acid (LPA) but with a higher affinity than PIP(2) (dissociation constant (K(d)) of 22 mum versus 39.5 mum for PIP(2)). More interestingly, unlike PIP(2), the association of villin with LPA inhibits all actin regulatory functions of villin. In addition, unlike PIP(2), LPA dramatically stimulates the tyrosine phosphorylation of villin by c-Src kinase. These studies suggest that in cells, selective interaction of villin with either PIP(2) or LPA could have dramatically different outcomes on actin reorganization as well as phospholipid-regulated cell signaling. These studies provide a novel regulatory mechanism for phospholipid-induced changes in the microfilament structure and cell function and suggest that LPA could be an intracellular regulator of the actin cytoskeleton.


Assuntos
Actinas/química , Lisofosfolipídeos/química , Proteínas dos Microfilamentos/química , Fosfatidilinositol 4,5-Difosfato/química , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Sítios de Ligação/fisiologia , Proteína Tirosina Quinase CSK , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lisofosfolipídeos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosforilação/fisiologia , Ligação Proteica/fisiologia , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Quinases da Família src
19.
J Biol Chem ; 283(33): 22709-22, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18482983

RESUMO

The ubiquitously expressed Src tyrosine kinases (c-Src, c-Yes, and c-Fyn) regulate intestinal cell growth and differentiation. Src activity is also elevated in the majority of malignant and premalignant tumors of the colon. The development of fibroblasts with the three ubiquitously expressed kinases deleted (SYF cells) has identified the role of Src proteins in the regulation of actin dynamics associated with increased cell migration and invasion. Despite this, unexpectedly nothing is known about the role of the individual Src kinases on intestinal cell cytoskeleton and/or cell migration. We have previously reported that villin, an epithelial cell-specific actin-modifying protein that regulates actin reorganization, cell morphology, cell migration, cell invasion, and apoptosis, is tyrosine-phosphorylated. In this report using the SYF cells reconstituted individually with c-Src, c-Yes, c-Fyn, and wild type or phosphorylation site mutants of villin, we demonstrate for the first time the absolute requirement for c-Src in villin-induced regulation of cell migration. The other major finding of our study is that contrary to previous reports, the nonreceptor tyrosine kinase, Jak3 (Janus kinase 3), does not regulate phosphorylation of villin or villin-induced cell migration and is, in fact, not expressed in intestinal epithelial cells. Further, we identify SHP-2 and PTP-PEST (protein-tyrosine phosphatase proline-, glutamate-, serine-, and threonine-rich sequence) as negative regulators of c-Src kinase and demonstrate a new function for these phosphatases in intestinal cell migration. Together, these data suggest that in colorectal carcinogenesis, elevation of c-Src or down-regulation of SHP-2 and/or PTP-PEST may promote cancer metastases and invasion by regulating villin-induced cell migration and cell invasion.


Assuntos
Movimento Celular/fisiologia , Proteínas Tirosina Quinases/metabolismo , Proteína Tirosina Quinase CSK , Células CACO-2/fisiologia , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/metabolismo , Células HeLa , Humanos , Janus Quinase 3/metabolismo , Invasividade Neoplásica , Fosforilação , Quinases da Família src
20.
Exp Cell Res ; 314(3): 530-42, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18054784

RESUMO

Autotaxin (ATX) is a potent tumor cell motogen that can produce lysophosphatidic acid (LPA) from lysophosphatidylcholine. LPA is a lipid mediator that has also been shown to modulate tumor cell invasion. Autotaxin mRNA is expressed at significant levels in the intestine. Likewise, LPA2 receptor levels have been shown to be elevated in colon cancers. The molecular mechanism of ATX/LPA-induced increase in intestinal cell migration however, remains poorly understood. Villin is an intestinal and renal epithelial cell specific actin regulatory protein that modifies epithelial cell migration. In this study we demonstrate that both Caco-2 (endogenous villin) and MDCK (exogenous villin) cells, which express primarily LPA2 receptors, show enhanced cell migration in response to ATX/LPA. ATX and LPA treatment results in the rapid formation of lamellipodia and redistribution of villin to these cell surface structures, suggesting a role for villin in regulating this initial event of cell locomotion. The LPA-induced increase in cell migration required activation of c-src kinase and downstream tyrosine phosphorylation of villin by c-src kinase. LPA stimulated cell motility was determined to be insensitive to pertussis toxin, but was regulated by activation of PLC-gamma 1. Together, our results show that in epithelial cells ATX and LPA act as strong stimulators of cell migration by recruiting PLC-gamma 1 and villin, both of which participate in the initiation of protrusion.


Assuntos
Movimento Celular/fisiologia , Mucosa Intestinal/metabolismo , Lisofosfolipídeos/fisiologia , Proteínas dos Microfilamentos/metabolismo , Complexos Multienzimáticos/fisiologia , Fosfodiesterase I/fisiologia , Pseudópodes/metabolismo , Pirofosfatases/fisiologia , Actinas/metabolismo , Animais , Células CACO-2 , Carcinoma/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Cães , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Neoplasias Intestinais/metabolismo , Lisofosfolipídeos/farmacologia , Proteínas dos Microfilamentos/efeitos dos fármacos , Proteínas dos Microfilamentos/farmacologia , Complexos Multienzimáticos/farmacologia , Fosfodiesterase I/farmacologia , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Diester Fosfórico Hidrolases , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Pseudópodes/efeitos dos fármacos , Pseudópodes/ultraestrutura , Pirofosfatases/farmacologia , Receptores de Ácidos Lisofosfatídicos/efeitos dos fármacos , Receptores de Ácidos Lisofosfatídicos/metabolismo , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...