Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 134(1)2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33277381

RESUMEN

Cell migration involves front-to-rear asymmetric focal adhesion (FA) dynamics, which facilitates trailing edge detachment and directional persistence. Here, we show that kindlin-2 is crucial for FA sliding and disassembly in migrating cells. Loss of kindlin-2 markedly reduced FA number and selectively impaired rear FA sliding and disassembly, resulting in defective rear retraction and reduced directional persistence during cell migration. Kindlin-2-deficient cells failed to develop serum-induced actomyosin-dependent tension at FAs. At the molecular level, kindlin-2 directly interacted with myosin light chain kinase (MYLK, hereafter referred to as MLCK), which was enhanced in response to serum stimulation. Serum deprivation inhibited rear FA disassembly, which was released in response to serum stimulation. Overexpression of the MLCK-binding kindlin-2 F0F1 fragment (amino acid residues 1-167), which inhibits the interaction of endogenous kindlin-2 with MLCK, phenocopied kindlin-2 deficiency-induced migration defects. Inhibition of MLCK, like loss of kindlin-2, also impaired trailing-edge detachment, rear FA disassembly and directional persistence. These results suggest a role of kindlin-2 in promoting actomyosin contractility at FAs, leading to increased rear FA sliding and disassembly, and directional persistence during cell migration.


Asunto(s)
Adhesiones Focales , Quinasa de Cadena Ligera de Miosina , Adhesión Celular , Movimiento Celular/genética , Adhesiones Focales/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosforilación
2.
Cell Mol Life Sci ; 80(1): 18, 2022 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-36564652

RESUMEN

Glomerular diseases afflict millions of people and impose an enormous burden on public healthcare costs worldwide. Identification of potential therapeutic targets for preventing glomerular diseases is of considerable clinical importance. CHILKBP is a focal adhesion protein and modulates a wide array of biological functions. However, little is known about the role of CHILKBP in glomerular diseases. To investigate the function of CHILKBP in maintaining the structure and function of podocytes in a physiologic setting, a mouse model (CHILKBP cKO) was generated in which CHILKBP gene was conditionally deleted in podocytes using the Cre-LoxP system. Ablation of CHILKBP in podocytes resulted in massive proteinuria and kidney failure in mice. Histologically, typical podocyte injury including podocyte loss, foot process effacement, and glomerulosclerosis was observed in CHILKBP cKO mice. Mechanistically, we identified ZO-1 as a key junctional protein that interacted with CHILKBP. Loss of CHILKBP in podocytes exhibited a significant reduction of ZO-1 expression, leading to abnormal actin organization, aberrant slit diaphragm protein expression and compromised podocyte filtration capacity. Restoration of CHILKBP or ZO-1 in CHILKBP-deficient podocytes effectively alleviated podocyte injury induced by the loss of CHILKBP in vitro and in vivo. Finally, we showed the glomerular expression of CHILKBP and ZO-1 was decreased in patients with proteinuric kidney diseases. Our findings reveal a novel signaling pathway consisting of CHILKBP and ZO-1 that plays an essential role in maintaining podocyte homeostasis and suggest novel therapeutic approaches to alleviate glomerular diseases.


Asunto(s)
Enfermedades Renales , Podocitos , Ratones , Animales , Podocitos/metabolismo , Glomérulos Renales/metabolismo , Enfermedades Renales/metabolismo , Transducción de Señal , Proteinuria/metabolismo
3.
Bioessays ; 43(9): e2100116, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34218442

RESUMEN

We propose a signaling pathway in which cell-extracellular matrix (ECM) adhesion components PINCH-1 and kindlin-2 sense mechanical signals from ECM and link them to proline biosynthesis, a vital metabolic pathway for macromolecule synthesis, redox balance, and ECM remodeling. ECM stiffening promotes PINCH-1 expression via integrin signaling, which suppresses dynamin-related protein 1 (DRP1) expression and mitochondrial fission, resulting in increased kindlin-2 translocation into mitochondria and interaction with Δ1 -pyrroline-5-carboxylate (P5C) reductase 1 (PYCR1). Kindlin-2 interaction with PYCR1 protects the latter from proteolytic degradation, leading to elevated PYCR1 level. Additionally, PINCH-1 promotes P5C synthase (P5CS) expression and P5C synthesis, which, together with increased PYCR1 level, support augmented proline biosynthesis. This signaling pathway is frequently activated in fibrosis and cancer, resulting in increased proline biosynthesis and excessive collagen matrix production, which in turn further promotes ECM stiffening. Targeting this signaling pathway, therefore, may provide an effective strategy for alleviating fibrosis and cancer progression.


Asunto(s)
Prolina , Pirrolina Carboxilato Reductasas , Matriz Extracelular , Dinámicas Mitocondriales , Pirrolina Carboxilato Reductasas/metabolismo , Transducción de Señal
4.
J Biol Chem ; 296: 100109, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33853759

RESUMEN

Cell-extracellular matrix (ECM) detachment is known to decrease extracellular signal-regulated kinase (ERK) signaling, an intracellular pathway that is central for control of cell behavior. How cell-ECM detachment is linked to downregulation of ERK signaling, however, is incompletely understood. We show here that focal adhesion protein Ras Suppressor 1 (RSU1) plays a critical role in cell-ECM detachment induced suppression of ERK signaling. We have identified prohibitin 2 (PHB2), a component of membrane lipid rafts, as a novel binding protein of RSU1, and mapped a major RSU1-binding site to PHB2 amino acids 150 to 206 in the C-terminal region of the PHB/SPFH (stomatin/prohibitin/flotillin/HflKC) domain. The PHB2 binding is mediated by multiple sites located in the N-terminal leucine-rich repeat region of RSU1. Depletion of PHB2 suppressed cell-ECM adhesion-induced ERK activation. Furthermore, cell-ECM detachment increased RSU1 association with membrane lipid rafts and interaction with PHB2. Finally, knockout of RSU1 or inhibition of RSU1 interaction with PHB2 by overexpression of the major RSU1-binding PHB2 fragment (amino acids 150-206) effectively suppressed the cell-ECM detachment induced downregulation of ERK signaling. Additionally, expression of venus-tagged wild-type RSU1 restored ERK signaling, while expression of venus-tagged PHB2-binding defective RSU1 mutant in which the N-terminal leucine-rich repeat region is deleted did not. Taken together, Our findings identify a novel RSU1-PHB2 signaling axis that senses cell-ECM detachment and links it to decreased ERK signaling.


Asunto(s)
Regulación hacia Abajo , Matriz Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Adhesión Celular/genética , Línea Celular Tumoral , Matriz Extracelular/genética , Humanos , Prohibitinas , Proteínas Represoras/genética
5.
Stem Cells ; 39(5): 650-668, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33529444

RESUMEN

Extracellular matrix (ECM) stiffness plays an important role in the decision making process of smooth muscle differentiation of mesenchymal stem cells (MSCs) but the underlying mechanisms are incompletely understood. Here we show that a signaling axis consisting of PINCH-1 and Notch2 is critically involved in mediating the effect of ECM stiffness on smooth muscle differentiation of MSCs. Notch2 level is markedly increased in ECM stiffness-induced smooth muscle differentiation of human placental MSCs. Knockdown of Notch2 from human placental MSCs effectively inhibits ECM stiffness-induced smooth muscle differentiation, whereas overexpression of North intracellular domain (NICD2) is sufficient to drive human placental MSC differentiation toward smooth muscle cells. At the molecular level, Notch2 directly interacts with PINCH-1. The interaction of Notch2 with PINCH-1 is significantly increased in response to ECM stiffness favoring smooth muscle differentiation. Furthermore, depletion of PINCH-1 from human placental MSCs reduces Notch2 level and consequently suppresses ECM stiffness-induced smooth muscle differentiation. Re-expression of PINCH-1, but not that of a Notch2-binding defective PINCH-1 mutant, in PINCH-1 knockdown human placental MSCs restores smooth muscle differentiation. Finally, overexpression of NICD2 is sufficient to override PINCH-1 deficiency-induced defect in smooth muscle differentiation. Our results identify an ECM stiffness-responsive PINCH-1-Notch2 interaction that is critically involved in ECM stiffness-induced smooth muscle differentiation of human placental MSCs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Matriz Extracelular/genética , Proteínas con Dominio LIM/genética , Músculo Liso/crecimiento & desarrollo , Receptor Notch2/genética , Diferenciación Celular/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Mecanotransducción Celular/genética , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Músculo Liso/metabolismo , Placenta/citología , Placenta/metabolismo , Placentación/genética , Embarazo , Transducción de Señal/genética
6.
Am J Respir Cell Mol Biol ; 65(1): 54-69, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33761308

RESUMEN

Pulmonary fibrosis is a progressive and fatal lung disease characterized by activation of lung fibroblasts and excessive deposition of collagen matrix. We show here that the concentrations of kindlin-2 and its binding partner PYCR1, a key enzyme for proline synthesis, are significantly increased in the lung tissues of human patients with pulmonary fibrosis. Treatment of human lung fibroblasts with TGF-ß1 markedly increased the expression of kindlin-2 and PYCR1, resulting in increased kindlin-2 mitochondrial translocation, formation of the kindlin-2-PYCR1 complex, and proline synthesis. The concentrations of the kindlin-2-PYCR1 complex and proline synthesis were markedly reduced in response to pirfenidone or nintedanib, two clinically approved therapeutic drugs for pulmonary fibrosis. Furthermore, depletion of kindlin-2 alone was sufficient to suppress TGF-ß1-induced increases of PYCR1 expression, proline synthesis, and fibroblast activation. Finally, using a bleomycin mouse model of pulmonary fibrosis, we show that ablation of kindlin-2 effectively reduced the concentrations of PYCR1, proline, and collagen matrix and alleviate the progression of pulmonary fibrosis in vivo. Our results suggest that kindlin-2 is a key promoter of lung fibroblast activation, collagen matrix synthesis, and pulmonary fibrosis, underscoring the therapeutic potential of targeting the kindlin-2 signaling pathway for control of this deadly lung disease.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Pulmón/metabolismo , Proteínas Musculares/metabolismo , Fibrosis Pulmonar/metabolismo , Transducción de Señal , Animales , Bleomicina/efectos adversos , Bleomicina/farmacología , Proteínas del Citoesqueleto/genética , Modelos Animales de Enfermedad , Fibroblastos/patología , Humanos , Pulmón/patología , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología
7.
Amino Acids ; 53(12): 1875-1890, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34283311

RESUMEN

Proline metabolic reprogramming is intimately involved in cancer progression. We recently identified a critical role of PINCH-1, a cell-extracellular matrix (ECM) adhesion protein whose expression is elevated in lung adenocarcinoma, in the promotion of proline biosynthesis, fibrosis and lung adenocarcinoma growth. How PINCH-1 promotes proline biosynthesis, however, was incompletely understood. In this study, we show that PINCH-1 promotes the expression of Δ1-pyrroline-5-carboxylate synthase (P5CS), a key enzyme that links glutamate metabolism to proline biosynthesis. Depletion of PINCH-1 from lung adenocarcinoma cells reduced the protein but not mRNA level of P5CS, resulting in down-regulation of the cellular level of P5C and cell proliferation. Treatment of the cells with protease inhibitor leupeptin effectively reversed PINCH-1 deficiency-induced reduction of the P5CS level. At the molecular level, PINCH-1, through its LIM2 domain, physically associated with P5CS in lung adenocarcinoma cells. Re-expression of wild type PINCH-1, but not that of the PINCH-1 LIM2 deletion mutant, in PINCH-1 deficient lung adenocarcinoma cells restored P5CS expression, proline biosynthesis and cell proliferation. Finally, P5CS expression, like that of PINCH-1, is elevated in human and mouse lung adenocarcinoma. Using a mouse model of lung adenocarcinoma in which PINCH-1 is conditionally ablated, we show that knockout of PINCH-1 from lung adenocarcinoma effectively reduced the P5CS level in vivo. Our results reveal an important role of PINCH-1 in the promotion of P5CS expression, which likely contributes to proline metabolic reprogramming and consequently lung adenocarcinoma progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma del Pulmón/metabolismo , Proteínas con Dominio LIM/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de la Membrana/metabolismo , Prolina/metabolismo , Células A549 , Animales , Línea Celular Tumoral , Proliferación Celular/fisiología , Regulación hacia Abajo/fisiología , Humanos , Ratones , Ratones Transgénicos , ARN Mensajero/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(35): 9349-9354, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28739949

RESUMEN

Kindlins and talins are integrin-binding proteins that are critically involved in integrin activation, an essential process for many fundamental cellular activities including cell-matrix adhesion, migration, and proliferation. As FERM-domain-containing proteins, talins and kindlins, respectively, bind different regions of ß-integrin cytoplasmic tails. However, compared with the extensively studied talin, little is known about how kindlins specifically interact with integrins and synergistically enhance their activation by talins. Here, we determined crystal structures of kindlin2 in the apo-form and the ß1- and ß3-integrin bound forms. The apo-structure shows an overall architecture distinct from talins. The complex structures reveal a unique integrin recognition mode of kindlins, which combines two binding motifs to provide specificity that is essential for integrin activation and signaling. Strikingly, our structures uncover an unexpected dimer formation of kindlins. Interrupting dimer formation impairs kindlin-mediated integrin activation. Collectively, the structural, biochemical, and cellular results provide mechanistic explanations that account for the effects of kindlins on integrin activation as well as for how kindlin mutations found in patients with Kindler syndrome and leukocyte-adhesion deficiency may impact integrin-mediated processes.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Integrinas/metabolismo , Proteínas Musculares/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Proteínas del Citoesqueleto/genética , Escherichia coli , Integrinas/genética , Modelos Moleculares , Proteínas Musculares/genética , Mutación , Unión Proteica , Conformación Proteica , Dominios Proteicos
9.
Stem Cells ; 35(2): 411-424, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27501743

RESUMEN

Aging deteriorates osteogenic capacity of mesenchymal stem/stromal cells (MSCs), contributing to imbalanced bone remodeling and osteoporosis. Glutaminase (Gls) catabolizes glutamine into glutamate at the first step of mitochondrial glutamine (Gln)-dependent anaplerosis which is essential for MSCs upon osteogenic differentiation. Estrogen-related receptor α (ERRα) regulates genes required for mitochondrial function. Here, we found that ERRα and Gls are upregulated by osteogenic induction in human MSCs (hMSCs). In contrast, osteogenic differentiation capacity and glutamine consumption of MSCs, as well as ERRα, Gls and osteogenic marker genes are significantly reduced with age. We demonstrated that ERRα binds to response elements on Gls promoter and affects glutamine anaplerosis through transcriptional induction of Gls. Conversely, mTOR inhibitor rapamycin, ERRα inverse agonist compound 29 or Gls inhibitor BPTES leads to reduced Gln anaplerosis and deteriorated osteogenic differentiation of hMSCs. Importantly, overexpression of ERRα or Gls restored impairment by these inhibitors. Finally, we proved that compensated ERRα or Gls expression indeed potentiated Gln anaplerosis and osteogenic capability of elderly mice MSCs in vitro. Together, we establish that Gls is a novel ERRα target gene and ERRα/Gls signaling pathway plays an important role in osteogenic differentiation of MSCs, providing new sights into novel regenerative therapeutics development. Our findings suggest that restoring age-related mitochondrial Gln-dependent anaplerosis may be beneficial for degenerative bone disorders such as osteoporosis. Stem Cells 2017;35:411-424.


Asunto(s)
Diferenciación Celular , Senescencia Celular , Glutaminasa/metabolismo , Glutamina/metabolismo , Células Madre Mesenquimatosas/citología , Mitocondrias/metabolismo , Osteogénesis , Receptores de Estrógenos/metabolismo , Envejecimiento/metabolismo , Animales , Resorción Ósea/patología , Calcificación Fisiológica/genética , Regulación de la Expresión Génica , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
10.
J Am Soc Nephrol ; 28(12): 3545-3562, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28775002

RESUMEN

Alteration of podocyte behavior is critically involved in the development and progression of many forms of human glomerular diseases. The molecular mechanisms that control podocyte behavior, however, are not well understood. Here, we investigated the role of Kindlin-2, a component of cell-matrix adhesions, in podocyte behavior in vivo Ablation of Kindlin-2 in podocytes resulted in alteration of actin cytoskeletal organization, reduction of the levels of slit diaphragm proteins, effacement of podocyte foot processes, and ultimately massive proteinuria and death due to kidney failure. Through proteomic analyses and in vitro coimmunoprecipitation experiments, we identified Rho GDP-dissociation inhibitor α (RhoGDIα) as a Kindlin-2-associated protein. Loss of Kindlin-2 in podocytes significantly reduced the expression of RhoGDIα and resulted in the dissociation of Rac1 from RhoGDIα, leading to Rac1 hyperactivation and increased motility of podocytes. Inhibition of Rac1 activation effectively suppressed podocyte motility and alleviated the podocyte defects and proteinuria induced by the loss of Kindlin-2 in vivo Our results identify a novel Kindlin-2-RhoGDIα-Rac1 signaling axis that is critical for regulation of podocyte structure and function in vivo and provide evidence that it may serve as a useful target for therapeutic control of podocyte injury and associated glomerular diseases.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Neuropéptidos/metabolismo , Podocitos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Inhibidor alfa de Disociación del Nucleótido Guanina rho/metabolismo , Albuminuria/metabolismo , Animales , Apoptosis , Movimiento Celular , Creatinina/análisis , Proteínas del Citoesqueleto/genética , Progresión de la Enfermedad , Femenino , Fibrosis , Genotipo , Humanos , Glomérulos Renales/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Proteínas de Neoplasias/genética , ARN Interferente Pequeño/metabolismo , Insuficiencia Renal/patología , Transducción de Señal
11.
Mol Cell ; 36(5): 819-30, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005845

RESUMEN

Integrin-linked kinase (ILK) plays a pivotal role in connecting transmembrane receptor integrin to the actin cytoskeleton and thereby regulating diverse cell-adhesion-dependent processes. The kinase domain (KD) of ILK is indispensable for its function, but the underlying molecular basis remains enigmatic. Here we present the crystal structure of the ILK KD bound to its cytoskeletal regulator, the C-terminal calponin homology domain of alpha-parvin. While maintaining a canonical kinase fold, the ILK KD displays a striking pseudoactive site conformation. We show that rather than performing the kinase function, this conformation specifically recognizes alpha-parvin for promoting effective assembly of ILK into focal adhesions. The alpha-parvin-bound ILK KD can simultaneously engage integrin beta cytoplasmic tails. These results thus define ILK as a distinct pseudokinase that mechanically couples integrin and alpha-parvin for mediating cell adhesion. They also highlight functional diversity of the kinase fold and its "active" site in mediating many biological processes.


Asunto(s)
Actinina/metabolismo , Adhesiones Focales/metabolismo , Proteínas Serina-Treonina Quinasas/química , Sitios de Unión , Cristalografía por Rayos X , Proteínas Quinasas Dependientes de AMP Cíclico/química , Humanos , Proteínas de Microfilamentos , Modelos Moleculares , Pliegue de Proteína , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína
12.
J Biol Chem ; 290(13): 8081-94, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25648889

RESUMEN

There is an intimate relationship between muscle and bone throughout life. However, how alterations in muscle functions in disease impact bone homeostasis is poorly understood. Amyotrophic lateral sclerosis (ALS) is a neuromuscular disease characterized by progressive muscle atrophy. In this study we analyzed the effects of ALS on bone using the well established G93A transgenic mouse model, which harbors an ALS-causing mutation in the gene encoding superoxide dismutase 1. We found that 4-month-old G93A mice with severe muscle atrophy had dramatically reduced trabecular and cortical bone mass compared with their sex-matched wild type (WT) control littermates. Mechanically, we found that multiple osteoblast properties, such as the formation of osteoprogenitors, activation of Akt and Erk1/2 pathways, and osteoblast differentiation capacity, were severely impaired in primary cultures and bones from G93A relative to WT mice; this could contribute to reduced bone formation in the mutant mice. Conversely, osteoclast formation and bone resorption were strikingly enhanced in primary bone marrow cultures and bones of G93A mice compared with WT mice. Furthermore, sclerostin and RANKL expression in osteocytes embedded in the bone matrix were greatly up-regulated, and ß-catenin was down-regulated in osteoblasts from G93A mice when compared with those of WT mice. Interestingly, calvarial bone that does not load and long bones from 2-month-old G93A mice without muscle atrophy displayed no detectable changes in parameters for osteoblast and osteoclast functions. Thus, for the first time to our knowledge, we have demonstrated that ALS causes abnormal bone remodeling and defined the underlying molecular and cellular mechanisms.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Remodelación Ósea , Atrofia Muscular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Esclerosis Amiotrófica Lateral/patología , Animales , Densidad Ósea , Células de la Médula Ósea/fisiología , Huesos/patología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Femenino , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/patología , Atrofia Muscular/patología , Mutación Missense , Osteoblastos/fisiología , Osteoclastos/fisiología , Ligando RANK/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
13.
J Cell Biochem ; 117(10): 2289-301, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26918647

RESUMEN

Cancer stem cells (CSCs) are capable of initiation and metastasis of tumors. Therefore, understanding the biology of CSCs and the interaction between CSCs and their counterpart non-stem cells is crucial for developing a novel cancer therapy. We used CSC-like and non-stem breast cancer MDA-MB-231 and MDA-MB-453 cells to investigate mammosphere formation. We investigated the role of the epithelial cadherin (E-cadherin)-extracellular signal-regulated kinase (Erk) axis in anoikis. Data from E-cadherin small hairpin RNA assay and mitogen-activated protein kinase kinase (MEK) inhibitor study show that activation of Erk, but not modulation of E-cadherin level, may play an important role in anoikis resistance. Next, the two cell subtypes were mixed and the interaction between them during mammosphere culture and xenograft tumor formation was investigated. Unlike CSC-like cells, increased secretion of interleukin-6 (IL-6) and growth-related oncogene (Gro) chemokines was detected during mammosphere culture in non-stem cells. Similar results were observed in mixed cells. Interestingly, CSC-like cells protected non-stem cells from anoikis and promoted tumor growth. Our results suggest bystander effects between CSC-like cells and non-stem cells. J. Cell. Biochem. 117: 2289-2301, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Anoicis/fisiología , Neoplasias de la Mama/patología , Efecto Espectador , Células Madre Neoplásicas/patología , Células Madre/patología , Animales , Antígenos CD , Western Blotting , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Células Madre/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Biol Chem ; 289(45): 31001-13, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25237194

RESUMEN

Integrin-mediated cell-extracellular matrix (ECM) adhesion is critical for control of intracellular signaling; however, the mechanisms underlying this "outside-in" signaling are incompletely understood. Here we show that depletion of kindlin-2 impairs integrin outside-in signaling. Kindlin-2 is tyrosine-phosphorylated upon cell-ECM adhesion. Furthermore, kindlin-2 binds Src in a cell-ECM adhesion-regulatable fashion. At the molecular level, the kindlin-2·Src interaction is mediated by the kindlin-2 F0 and the Src SH2 and SH3 domains. Src activation increases kindlin-2 tyrosine phosphorylation and the kindlin-2·Src interaction. Conversely, inhibition of Src reduces kindlin-2 tyrosine phosphorylation and diminishes the kindlin-2·Src interaction. Finally, disruption of the kindlin-2·Src interaction, unlike depletion of kindlin-2, impairs neither cell-ECM adhesion nor cell-ECM adhesion-induced focal adhesion kinase Tyr-397 phosphorylation. However, it markedly inhibits cell-ECM adhesion-induced paxillin tyrosine phosphorylation, cell migration, and proliferation. These results suggest that kindlin-2 tyrosine phosphorylation and interaction with Src serve as a regulatable switch downstream of focal adhesion kinase in the integrin outside-in signaling circuit, relaying signals from cell-ECM adhesion to paxillin that control cell migration and proliferation.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Integrinas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Familia-src Quinasas/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Adhesión Celular , Movimiento Celular , Proliferación Celular , Matriz Extracelular/metabolismo , Fibroblastos/citología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación de la Expresión Génica , Humanos , Glomérulos Renales/metabolismo , Ratones , Paxillin/metabolismo , Fosforilación , Podocitos/citología , Unión Proteica , Proteínas Recombinantes/metabolismo , Transducción de Señal , Tirosina , Dominios Homologos src
15.
J Transl Med ; 13: 321, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26444413

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is a highly lethal cancer, and its underlying molecular mechanisms are poorly understood. Recent large-scale genome-wide association studies in Chinese Han populations have identified an ESCC susceptibility locus within the SLC39A6 gene. Here, we sought to explore the expression and biological function of SLC39A6 in ESCC. METHODS: Multiethnic validation of SLC39A6 protein expression was performed in different cohorts of patients from Chinese Han and Kazakh populations in the Xinjiang region by immunohistochemistry. The associations among SLC39A6 expression, clinicopathological parameters, and prognosis outcomes of ESCC were analyzed. And the effects of SLC39A6 silencing by siRNA on cell proliferation, apoptosis, and invasiveness, as well as the proteins involved in epithelial-to-mesenchymal transition (EMT) of esophageal cancer cells, were studied. RESULTS: SLC39A6 protein expression increased progressively from normal esophageal epithelium (NEE) to low-grade intraepithelial neoplasia to ESCC, and finally reached the highest in high-grade intraepithelial neoplasia from Han ethnic. Similarly, SLC39A6 protein was significantly overexpressed in Kazakh ethnic ESCC compared with that in NEE. Increased expression of SLC39A6 was found to be closely correlated with histological grade and early Tumor-Node-Metastasis stage I/II. High tumorous SLC39A6 expression was significantly correlated with shorter overall survival (OS). Cox regression analysis confirmed that SLC39A6 expression was an independent prognostic factor for poor OS in ESCC. Experimentally, the suppression of SLC39A6 expression promoted ESCC cell apoptosis but abrogated proliferation and invasion, and induced an EMT phenotype that included enhanced expression of E-cadherin, loss of vimentin, and morphological changes in ESCC cells in vitro. CONCLUSIONS: Combined, our findings highlight a tumor-promoting role for SLC39A6 in ESCC, suggesting that SLC39A6 could serve as an early detector of high-risk subjects and prognostic biomarker. The targeting of SLC39A6 might be a potential therapeutic strategy for blocking ESCC.


Asunto(s)
Proteínas de Transporte de Catión/genética , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/terapia , Proteínas de Neoplasias/genética , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Biomarcadores de Tumor/metabolismo , Carcinoma/diagnóstico , Carcinoma/etnología , Carcinoma/metabolismo , Carcinoma/terapia , Proliferación Celular , China , Estudios de Cohortes , Transición Epitelial-Mesenquimal , Neoplasias Esofágicas/etnología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Fenotipo , Pronóstico , Modelos de Riesgos Proporcionales , ARN Interferente Pequeño/metabolismo , Análisis de Matrices Tisulares , Resultado del Tratamiento , Regulación hacia Arriba
16.
J Biol Chem ; 288(39): 28021-33, 2013 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-23897811

RESUMEN

Aberrant expression of histone deacetylases (HDACs) is associated with carcinogenesis. Some HDAC inhibitors are widely considered as promising anticancer therapeutics. A major obstacle for development of HDAC inhibitors as highly safe and effective anticancer therapeutics is that our current knowledge on the contributions of different HDACs in various cancer types remains scant. Here we report that the expression level of HDAC10 was significantly lower in patients exhibiting lymph node metastasis compared with that in patients lacking lymph node metastasis in human cervical squamous cell carcinoma. Forced expression of HDAC10 in cervical cancer cells significantly inhibited cell motility and invasiveness in vitro and metastasis in vivo. Mechanistically, HDAC10 suppresses expression of matrix metalloproteinase (MMP) 2 and 9 genes, which are known to be critical for cancer cell invasion and metastasis. At the molecular level, HDAC10 binds to MMP2 and -9 promoter regions, reduces the histone acetylation level, and inhibits the binding of RNA polymerase II to these regions. Furthermore, an HDAC10 mutant lacking histone deacetylase activity failed to mimic the functions of full-length protein. These results identify a critical role of HDAC10 in suppression of cervical cancer metastasis, underscoring the importance of developing isoform-specific HDAC inhibitors for treatment of certain cancer types such as cervical squamous cell carcinoma.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Adulto , Anciano , Animales , Movimiento Celular , Inhibidores Enzimáticos/farmacología , Femenino , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Metástasis de la Neoplasia , Regiones Promotoras Genéticas , Isoformas de Proteínas/metabolismo , Interferencia de ARN , Neoplasias del Cuello Uterino/patología
17.
Acta Pharmacol Sin ; 35(9): 1199-206, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25152024

RESUMEN

AIM: Mig-2 (also known as Kindlin-2 and FERMT2) is an important regulator of integrin activation and cell-extracellular matrix adhesion, and involved in carcinogenesis and tumor progression. The aim of this study was to investigate the role of mig-2 in cisplatin-induced apoptosis of human glioma cells in vitro. METHODS: The expression of mig-2 was modulated in human glioma H4, HS 683 and U-87 MG cells by transfection with a plasmid carrying mig-2 or mig-2 siRNA. Cisplatin-induced apoptosis was detected using Annexin V/PI staining and flow cytometry, as well as MTS analyses. The expression of apoptosis-related or signaling proteins was examined using Western blotting analysis. H4 cells were transfected with plasmids carrying mig-2 mutants to determine the functional domain of mig-2. RESULTS: In the 3 glioma cell lines tested, overexpression of mig-2 significantly attenuated cisplatin-induced apoptosis, whereas knock-down of mig-2 potentiated the apoptosis. The mechanisms of action of mig-2 were further addressed in H4 cells: overexpression of mig-2 markedly reduced cleaved caspase-9, caspase-8, caspase-3 and PARP, as well as p-JNK and p-p38, and increased p-AKT in cisplatin-treated H4 cells, whereas mig-2 siRNA reversely changed these apoptosis-related and signaling proteins. Furthermore, pretreatment with JNK inhibitor SP600125 and p38 inhibitor SB203580, or with AKT inhibitor LY294002 abolished the effects of mig-2 on cisplaxtin-induced apoptosis. In H4 cells, GFP-mig-2 F3 plasmid that contained only the F3 subdomain showed the same efficiency in attenuating cisplatin-induced apoptosis, as the mig-2 wild-type vector did, whereas GFP-mig-2 (1-541) plasmid that lacked the F3 subdomain was inactive. CONCLUSION: Mig-2 significantly attenuates the antitumor action of cisplatin against human glioma cells in vitro through AKT/JNK and AKT/p38 signaling pathways. The F3 subdomain of mig-2 is necessary and sufficient for this effect.


Asunto(s)
Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Glioma/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Línea Celular Tumoral , Humanos , Transducción de Señal/efectos de los fármacos
18.
J Biol Chem ; 287(39): 32394-405, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22843679

RESUMEN

Migfilin is critical for cell shape and motile regulation. However, its pathological role in glioma is unknown. Using an immunohistochemical staining assay, we demonstrate that there is a significant correlation between expression of Migfilin and pathological tumor grade in 217 clinical glioma samples. High Migfilin expression is associated with poor prognosis for patients with glioma. Investigation of the molecular mechanism shows that Migfilin promotes migration and invasion in glioma cells. Moreover, Migfilin positively modulates the expression and activity of epidermal growth factor receptor, and Migfilin-mediated migration and invasion depend on epidermal growth factor receptor-induced PLC-γ and STAT3-signaling pathways. Our results may provide significant clinical application, including use of Migfilin as a molecular marker in glioma for early diagnosis and as an indicator of prognosis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular , Proteínas del Citoesqueleto/metabolismo , Receptores ErbB/biosíntesis , Glioma/metabolismo , Fosfolipasa C gamma/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Biomarcadores de Tumor/genética , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Proteínas del Citoesqueleto/genética , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioma/genética , Glioma/patología , Humanos , Invasividad Neoplásica , Fosfolipasa C gamma/genética , Factor de Transcripción STAT3/genética
19.
J Biol Chem ; 287(25): 21450-60, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22556421

RESUMEN

Bone remodeling is a complex process that must be precisely controlled to maintain a healthy life. We show here that filamin-binding LIM protein 1 (FBLP-1, also known as migfilin), a kindlin- and filamin-binding focal adhesion protein, is essential for proper control of bone remodeling. Genetic inactivation of FBLIM1 (the gene encoding FBLP-1) in mice resulted in a severe osteopenic phenotype. Primary FBLP-1 null bone marrow stromal cells (BMSCs) exhibited significantly reduced extracellular matrix adhesion and migration compared with wild type BMSCs. Loss of FBLP-1 significantly impaired the growth and survival of BMSCs in vitro and decreased the number of osteoblast (OB) progenitors in bone marrow and OB differentiation in vivo. Furthermore, the loss of FBLP-1 caused a dramatic increase of osteoclast (OCL) differentiation in vivo. The level of receptor activator of nuclear factor κB ligand (RANKL), a key regulator of OCL differentiation, was markedly increased in FBLP-1 null BMSCs. The capacity of FBLP-1 null bone marrow monocytes (BMMs) to differentiate into multinucleated OCLs in response to exogenously supplied RANKL, however, was not different from that of WT BMMs. Finally, we show that a loss of FBLP-1 promotes activating phosphorylation of ERK1/2. Inhibition of ERK1/2 activation substantially suppressed the increase of RANKL induced by the loss of FBLP-1. Our results identify FBLP-1 as a key regulator of bone homeostasis and suggest that FBLP-1 functions in this process through modulating both the intrinsic properties of OB/BMSCs (i.e., BMSC-extracellular matrix adhesion and migration, cell growth, survival, and differentiation) and the communication between OB/BMSCs and BMMs (i.e., RANKL expression) that controls osteoclastogenesis.


Asunto(s)
Remodelación Ósea/fisiología , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/genética , Movimiento Celular/fisiología , Proteínas del Citoesqueleto/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Monocitos/citología , Monocitos/metabolismo , Osteoblastos/citología , Osteoclastos/citología , Fosforilación/fisiología , Células del Estroma/citología , Células del Estroma/metabolismo
20.
J Cell Sci ; 124(Pt 6): 879-91, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21325030

RESUMEN

Kindlin-2 is a FERM and PH domain-containing integrin-binding protein that is emerging as an important regulator of integrin activation. How kindlin-2 functions in integrin activation, however, is not known. We report here that kindlin-2 interacts with multiple phosphoinositides, preferentially with phosphatidylinositol 3,4,5-trisphosphate. Although integrin-binding is essential for focal adhesion localization of kindlin-2, phosphoinositide-binding is not required for this process. Using biologically and clinically relevant glomerular podocytes as a model system, we show that integrin activation and dependent processes are tightly regulated by kindlin-2: depletion of kindlin-2 reduced integrin activation, matrix adhesion and fibronectin matrix deposition, whereas overexpression of kindlin-2 promoted these processes. Furthermore, we provide evidence showing that kindlin-2 is involved in phosphoinositide-3-kinase-mediated regulation of podocyte-matrix adhesion and fibronectin matrix deposition. Mechanistically, kindlin-2 promotes integrin activation and integrin-dependent processes through interacting with both integrins and phosphoinositides. TGF-ß1, a mediator of progressive glomerular failure, markedly increased the level of kindlin-2 and fibronectin matrix deposition, and the latter process was reversed by depletion of kindlin-2. Our results reveal important functions of kindlin-2 in the regulation of podocyte-matrix adhesion and matrix deposition and shed new light on the mechanism whereby kindlin-2 functions in these processes.


Asunto(s)
Fibronectinas/metabolismo , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfatidilinositoles/metabolismo , Podocitos/citología , Podocitos/metabolismo , Adhesión Celular , Línea Celular , Matriz Extracelular/química , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibronectinas/genética , Humanos , Integrina beta1/genética , Integrina beta3/genética , Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Podocitos/química , Unión Proteica , Estructura Terciaria de Proteína , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA