Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(49): 24600-24609, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31740618

RESUMEN

Cell adhesion is essential for proper tissue architecture and function in multicellular organisms. Cell adhesion molecules not only maintain tissue integrity but also possess signaling properties that contribute to diverse cellular events such as cell growth, survival, differentiation, polarity, and migration; however, the underlying molecular basis remains poorly defined. Here we identify that the cell adhesion signal initiated by the tight-junction protein claudin-6 (CLDN6) regulates nuclear receptor activity. We show that CLDN6 recruits and activates Src-family kinases (SFKs) in second extracellular domain-dependent and Y196/200-dependent manners, and SFKs in turn phosphorylate CLDN6 at Y196/200. We demonstrate that the CLDN6/SFK/PI3K/AKT axis targets the AKT phosphorylation sites in the retinoic acid receptor γ (RARγ) and the estrogen receptor α (ERα) and stimulates their activities. Interestingly, these phosphorylation motifs are conserved in 14 of 48 members of human nuclear receptors. We propose that a similar link between diverse cell adhesion and nuclear receptor signalings coordinates a wide variety of physiological and pathological processes.


Asunto(s)
Adhesión Celular/fisiología , Claudinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Línea Celular , Claudinas/genética , Receptor alfa de Estrógeno/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Dominios Proteicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Tirosina/genética , Familia-src Quinasas/metabolismo , Receptor de Ácido Retinoico gamma
2.
J Cell Physiol ; 236(10): 7134-7143, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33694168

RESUMEN

The neurovascular unit (NVU) consists of neurons, glial cells, microvascular cells, and extracellular matrix, and is involved in a variety of physiological and pathological processes in the central nervous system (CNS). Within the NVU, the microvascular endothelial cells and pericytes principally contribute to maintaining the integrity of the blood-brain barrier (BBB). Various types of cells are connected to each other in the NVU by diverse cell adhesion molecules, of which claudin-5 (CLDN5) is by far the most abundantly expressed tight-junction protein in brain microvascular endothelial cells and absolutely required for the maintenance of the BBB. This review highlights recent progress in understanding the region-specific regulation and dysregulation of CLDN5 expression in CNS health and disorders. We also discuss how CLDN5 expression is regionally disrupted within the NVU. In addition, we focus on the link between cell adhesion and transcription factor signalings and describe the possible involvement of CLDN5-adhesion signaling in brain health and disorders.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalopatías/metabolismo , Claudina-5/metabolismo , Células Endoteliales/metabolismo , Transducción de Señal , Uniones Estrechas/metabolismo , Animales , Barrera Hematoencefálica/patología , Encefalopatías/genética , Encefalopatías/patología , Claudina-5/genética , Células Endoteliales/patología , Regulación de la Expresión Génica , Humanos , Uniones Estrechas/genética , Uniones Estrechas/patología
3.
Int J Mol Sci ; 22(1)2020 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-33383868

RESUMEN

We previously reported that site-selective claudin-5 (CLDN5) breakdown and protein kinase A (PKA) activation are observed in brain microvessels of schizophrenia, but the underlying molecular basis remains unknown. The 5-HT1 receptors decline the intracellular cAMP levels and inactivate the major downstream PKA, and the 5-HT1A receptor is a promising target for schizophrenia. Therefore, we elucidated the involvement of serotonin/5-HT1A signaling in the endothelial CLDN5 expression. We demonstrate, by immunohistochemistry using post-mortem human brain tissue, that the 5-HT1A receptor is expressed in brain microvascular endothelial cells (BMVECs) and mural cells of the normal prefrontal cortex (PFC) gray matter. We also show that PKA is aberrantly activated not only in BMVECs but also in mural cells of the schizophrenic PFC. We subsequently revealed that the endothelial cell-pericyte tube-like structure was formed in a novel two-dimensional co-culture of human primary BMVECs and a human brain-derived pericyte cell line, in both of which the 5-HT1A receptor was expressed. Furthermore, we disclose that the serotonin/5-HT1A signaling enhances endothelial CLDN5 expression in BMVECs under two-dimensional co-culture conditions. Our findings provide novel insights into the physiological and pathological significance of serotonin/5-HT1A signaling in the region-specific regulation of the blood-brain barrier.


Asunto(s)
Claudina-5/genética , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Acoplamiento Neurovascular , Receptor de Serotonina 5-HT1A/metabolismo , Serotonina/metabolismo , Transducción de Señal , Biomarcadores , Encéfalo/metabolismo , Comunicación Celular , Claudina-5/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Pericitos/patología
4.
Hippocampus ; 29(6): 539-549, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30365199

RESUMEN

Neural progenitors acquire GFAP expression during the perinatal period and continue to generate granule cells (GCs) in the hippocampal dentate gyrus throughout adulthood. Cellular characteristics of GFAP+ progenitor-derived late-born GCs in comparison with early-born GCs remain unknown. Using genetic fate mapping in mice, we show that early- and late-born GCs are concentrated in the outer and inner side of the GC layer, respectively. We then identify that a nuclear orphan receptor Nr4A2 is preferentially expressed by early-born GCs. Nr4a2 expression is dynamically regulated in response to restraint stress and glucocorticoid levels, indicating that Nr4a2 is a stress-regulated gene in GCs. Acute stress suppresses but chronic stress conversely induces Nr4a2 expression in GCs. The survival of newly generated GCs is impaired by chronic restraint stress and long-term stress after middle age decreases the proportion of late-born GCs in aged mice. Thus, early- and late-born GCs exhibit characteristic anatomical distribution, differential gene expression, and distinct response to environmental stress.


Asunto(s)
Giro Dentado/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Giro Dentado/citología , Giro Dentado/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Glucocorticoides/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neurogénesis/fisiología , Restricción Física , Estrés Fisiológico
5.
Molecules ; 22(10)2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-28994732

RESUMEN

Due to their lower production cost compared with monoclonal antibodies, single-chain variable fragments (scFvs) have potential for use in several applications, such as for diagnosis and treatment of a range of diseases, and as sensor elements. However, the usefulness of scFvs is limited by inhomogeneity through the formation of dimers, trimers, and larger oligomers. The scFv protein is assumed to be in equilibrium between the closed and open states formed by assembly or disassembly of VH and VL domains. Therefore, the production of an scFv with equilibrium biased to the closed state would be critical to overcome the problem in inhomogeneity of scFv for industrial or therapeutic applications. In this study, we obtained scFv clones stable against GA-pyridine, an advanced glycation end-product (AGE), by using a combination of a phage display system and random mutagenesis. Executing the bio-panning at 37 °C markedly improved the stability of scFvs. We further evaluated the radius of gyration by small-angle X-ray scattering (SAXS), obtained compact clones, and also visualized open.


Asunto(s)
Productos Finales de Glicación Avanzada/inmunología , Compuestos de Piridinio/inmunología , Anticuerpos de Cadena Única/biosíntesis , Secuencia de Aminoácidos , Biblioteca de Péptidos , Dominios Proteicos , Multimerización de Proteína , Estabilidad Proteica , Anticuerpos de Cadena Única/química
6.
Am J Pathol ; 184(6): 1683-94, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24720953

RESUMEN

Laminin α1 (LAMA1), a subunit of the laminin-111 basement membrane component, has been implicated in various biological functions in vivo and in vitro. Although LAMA1 is present in kidney, its roles in the kidney are unknown because of early embryonic lethality. Herein, we used a viable conditional knockout mouse model with a deletion of Lama1 in the epiblast lineage (Lama1(CKO)) to study the role of LAMA1 in kidney development and function. Adult Lama1(CKO) mice developed focal glomerulosclerosis and proteinuria with age. In addition, mesangial cell proliferation was increased, and the mesangial matrix, which normally contains laminin-111, was greatly expanded. In vitro, mesangial cells from Lama1(CKO) mice exhibited significantly increased proliferation compared with those from controls. This increased proliferation was inhibited by the addition of exogenous LAMA1-containing laminin-111, but not by laminin-211 or laminin-511, suggesting a specific role for LAMA1 in regulating mesangial cell behavior. Moreover, the absence of LAMA1 increased transforming growth factor (TGF)-ß1-induced Smad2 phosphorylation, and inhibitors of TGF-ß1 receptor I kinase blocked Smad2 phosphorylation in both control and Lama1(CKO) mesangial cells, indicating that the increased Smad2 phosphorylation occurred in the absence of LAMA1 via the TGF-ß1 receptor. These findings suggest that LAMA1 plays a critical role in kidney function and kidney aging by regulating the mesangial cell population and mesangial matrix deposition through TGF-ß/Smad signaling.


Asunto(s)
Envejecimiento/metabolismo , Proliferación Celular , Matriz Extracelular/metabolismo , Mesangio Glomerular/metabolismo , Laminina/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Animales , Matriz Extracelular/genética , Matriz Extracelular/patología , Mesangio Glomerular/patología , Glomerulonefritis/genética , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Laminina/genética , Ratones , Ratones Noqueados , Fosforilación/genética , Proteinuria/genética , Proteinuria/metabolismo , Proteinuria/patología , Transducción de Señal/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
7.
BMC Cancer ; 15: 53, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25884418

RESUMEN

BACKGROUND: S100 family proteins have recently been identified as biomarkers in various cancers. Of this protein family, S100A14 and S100A16 are also believed to play an important role in tumor progression. The aim of the present study was to clarify the clinical significance and functional role of these molecules in breast cancer. METHODS: In a clinical study, an immunohistochemical analysis of S100A14 and S100A16 expression in archival specimens of primary tumors of 167 breast cancer patients was performed. The relationship of S100A14 and S100A16 expression to patient survival and clinicopathological variables was statistically analyzed. In an experimental study, the subcellular localization and function of these molecules was examined by using the human breast cancer cell lines MCF7 and SK-BR-3, both of which highly express S100A14 and S100A16 proteins. Cells transfected with expression vectors and siRNA for these genes were characterized using in vitro assays for cancer invasion and metastasis. RESULTS: Immunohistochemical analysis of 167 breast cancer cases showed strong cell membrane staining of S100A14 (53% of cases) and S100A16 (31% of cases) with a significant number of cases with co-expression (p < 0.001). Higher expression levels of these proteins were significantly associated with a younger age (<60 years), ER-negative status, HER2-positive status and a poorer prognosis. Co-expression of the two proteins showed more aggressive features with poorer prognosis. In the human breast cancer cell lines MCF7 and SK-BR-3, both proteins were colocalized on the cell membrane mainly at cell-cell attachment sites. Immunoprecipitation and immunofluorescence analyses demonstrated that the 100A14 protein can bind to actin localized on the cell membrane in a calcium-independent manner. A Boyden chamber assay showed that S100A14 and S100A16 knockdown substantially suppressed the invasive activity of both cell lines. Cell motility was also inhibited by S100A14 knockdown in a modified dual color wound-healing assay. CONCLUSIONS: To our knowledge, this is the first report showing the correlation of expression of S100A14, S100A16, and co-expression of these proteins with poor prognosis of breast cancer patients. In addition, our findings indicate that S100A14 and S100A16 can promote invasive activity of breast cancer cells via an interaction with cytoskeletal dynamics. S100A14 and S100A16 might be prognostic biomarkers and potential therapeutic targets for breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Proteínas de Unión al Calcio/genética , Expresión Génica , Proteínas S100/genética , Actinas/metabolismo , Adulto , Anciano , Biomarcadores de Tumor , Neoplasias de la Mama/patología , Neoplasias de la Mama/cirugía , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Unión Proteica , Transporte de Proteínas , Proteínas S100/metabolismo
8.
Cells ; 12(13)2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37443730

RESUMEN

Cell adhesion molecules, including integrins, cadherins, and claudins (CLDNs), are known to activate Src-family kinases (SFKs) that organize a variety of physiological and pathological processes; however, the underlying molecular basis remains unclear. Here, we identify the SFK members that are coupled with the CLDN6-adhesion signaling. Among SFK subtypes, BLK, FGR, HCK, and SRC were highly expressed in F9 cells and concentrated with CLDN6 along cell borders during epithelial differentiation. Immunoprecipitation assay showed that BLK and SRC, but not FGR or HCK, form a complex with CLDN6 via the C-terminal cytoplasmic domain. We also demonstrated, by pull-down assay, that recombinant BLK and SRC proteins directly bind to the C-terminal cytoplasmic domain of CLDN6 (CLDN6C). Unexpectedly, both recombinant SFK proteins recognized the CLDN6C peptide in a phosphotyrosine-independent manner. Furthermore, by comparing phenotypes of F9:Cldn6:Blk-/- and F9:Cldn6:Src-/- cells with those of wild-type F9 and F9:Cldn6 cells, we revealed that BLK and SRC are essential for CLDN6-triggered cellular events, namely epithelial differentiation and the expression of retinoid acid receptor target genes. These results indicate that selective SFK members appear to participate in the CLDN-adhesion signaling.


Asunto(s)
Transducción de Señal , Familia-src Quinasas , Familia-src Quinasas/metabolismo , Moléculas de Adhesión Celular , Integrinas , Receptores de Ácido Retinoico , Claudinas/genética , Claudinas/metabolismo
9.
J Biomed Biotechnol ; 2011: 253048, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22162632

RESUMEN

Tight junctions are intercellular junctions localized at the most apical end of the lateral plasma membrane. They consist of four kinds of transmembrane proteins (occludin, claudins, junctional adhesion molecules, and tricellulin) and huge numbers of scaffolding proteins and contribute to the paracellular barrier and fence function. The mutation and deletion of these proteins impair the functions of tight junctions and cause various human diseases. In this paper, we provide an overview of recent studies on transmembrane proteins of tight junctions and highlight the functional significance of tight junctions, extracellular matrix, and nuclear receptors in epithelial differentiation.


Asunto(s)
Células Epiteliales/citología , Matriz Extracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Uniones Estrechas/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Claudinas/genética , Claudinas/metabolismo , Eliminación de Gen , Humanos , Moléculas de Adhesión de Unión , Proteína 2 con Dominio MARVEL , Proteínas de la Membrana/genética , Mutación/genética , Ocludina
10.
Biomedicines ; 9(3)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33801826

RESUMEN

Junctional adhesion molecules (JAMs) are expressed in diverse types of stem and progenitor cells, but their physiological significance has yet to be established. Here, we report that JAMs exhibit a novel mode of interaction and biological activity in adipose-derived stromal/stem cells (ADSCs). Among the JAM family members, JAM-B and JAM-C were concentrated along the cell membranes of mouse ADSCs. JAM-C but not JAM-B was broadly distributed in the interstitial spaces of mouse adipose tissue. Interestingly, the JAM-C ectodomain was cleaved and secreted as a soluble form (sJAM-C) in vitro and in vivo, leading to deposition in the fat interstitial tissue. When ADSCs were grown in culture plates coated with sJAM-C, cell adhesion, cell proliferation and the expression of five mesenchymal stem cell markers, Cd44, Cd105, Cd140a, Cd166 and Sca-1, were significantly elevated. Moreover, immunoprecipitation assay showed that sJAM-C formed a complex with JAM-B. Using CRISPR/Cas9-based genome editing, we also demonstrated that sJAM-C was coupled with JAM-B to stimulate ADSC adhesion and maintenance. Together, these findings provide insight into the unique function of sJAM-C in ADSCs. We propose that JAMs contribute not only to cell-cell adhesion, but also to cell-matrix adhesion, by excising their ectodomain and functioning as a niche-like microenvironment for stem and progenitor cells.

11.
Sci Rep ; 11(1): 16372, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34385585

RESUMEN

EMI Domain Containing 1 (EMID1) was identified as a potential candidate metastasis-promoting gene. We sought to clarify the molecular function of EMID1 and the protein expression. Overexpression and knockdown studies using mouse tumor cell lines identified two novel functions of EMID1: intracellular signaling involving enhancement of cell growth via cell cycle promotion and suppression of cell motility, and inhibition of cell-matrix adhesion by extracellularly secreted EMID1. EMID1 deposited on the culture dish induced self-detachment of cells that overexpressed the protein and inhibited adhesion of additionally seeded cells. This multifunctional property involving both intracellular signaling and the extracellular matrix suggests that EMID1 may be a matricellular proteins. Expression analysis using immunohistochemical staining revealed expression of EMID1 that was limited to chief cells of the gastric fundic gland and ß cells of the pancreatic islets in normal adult human tissues, implying cell-specific functions of this molecule. In addition, increased expression of EMID1 protein detected in some cases of human cancers implies that EMID1 might be a new therapeutic target for cancer treatment.


Asunto(s)
Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Transducción de Señal/genética , Animales , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Matriz Extracelular/genética , Matriz Extracelular/patología , Fundus Gástrico/patología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Células Secretoras de Insulina/patología , Ratones , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Procesos Neoplásicos
12.
Mol Cancer Res ; 19(7): 1208-1220, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33727343

RESUMEN

Cell adhesion proteins not only maintain tissue integrity, but also possess signaling abilities to organize diverse cellular events in a variety of physiologic and pathologic processes; however, the underlying mechanism remains obscure. Among cell adhesion molecules, the claudin (CLDN) family is often aberrantly expressed in various cancers, but the biological relevance and molecular basis for this observation have not yet been established. Here, we show that high CLDN6 expression accelerates cellular proliferation and migration in two distinct human endometrial cancer cell lines in vitro. Using a xenograft model, we also revealed that aberrant CLDN6 expression promotes tumor growth and invasion in endometrial cancer tissues. The second extracellular domain and Y196/200 of CLDN6 were required to recruit and activate Src-family kinases (SFK) and to stimulate malignant phenotypes. Knockout and overexpression of ESR1 in endometrial carcinoma cells showed that the CLDN6-adhesion signal links to estrogen receptor α (ERα) to advance tumor progression. In particular, aberrant CLDN6-ERα signaling contributed to collective cell behaviors in the leading front of endometrial cancer cells. Importantly, we demonstrate that CLDN6/SFK/PI3K-dependent AKT and SGK (serum- and glucocorticoid-regulated kinase) signaling in endometrial cancer cells targets Ser518 in the human ERα to activate ERα transcriptional activity in a ligand-independent manner, thereby promoting tumor progression. Furthermore, CLDN6, at least in part, also regulated gene expression in an ERα-independent manner. IMPLICATIONS: The identification of this machinery highlights regulation of the transcription factors by cell adhesion to advance tumor progression.


Asunto(s)
Adhesión Celular/genética , Claudinas/genética , Neoplasias Endometriales/genética , Receptor alfa de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica , Transducción de Señal/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Claudinas/metabolismo , Progresión de la Enfermedad , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Receptor alfa de Estrógeno/metabolismo , Femenino , Células HEK293 , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Ratones Endogámicos NOD , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trasplante Heterólogo , Familia-src Quinasas/metabolismo
13.
Clin Exp Metastasis ; 36(4): 411-422, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31263990

RESUMEN

Cancer metastasis shows great diversity in target organs, routes and molecular mechanisms depending on the type of cancer and even on the individual patients. To identify key molecules involved in metastasis, we constructed a murine model system including multiple sublines with different organotropism and pathways of metastasis. We selected metastatic sublines from a murine mammary tumor cell line MCH66. Using this model, we extracted metastasis-related molecules by gene expression screening methods and verified their metastasis-promoting effects by gene knockdown or overexpression experiments. For the candidates promoting metastasis, we analyzed molecular functions involved in metastasis: cell growth, motility and invasive activity. We established a metastasis model including low metastatic sublines (66C8, 66LM, 66-4) and highly metastatic counterparts with various organotropism, such as to the lung (66Lu10), liver (HM-KAN5) and general organs (66HM and its clones: HM1-6 and HM1-7). The sublines basically exhibited the invasion-independent metastasis pathway characterized by endothelial cell-covered tumor emboli, whereas 66HM and HM-KAN5 showed an alternative metastasis pathway based on invasion in part and in whole, respectively. Comprehensive gene analysis extracted several molecular candidates responsible for metastasis. S100A14 was identified as one of the promissing candidates promoting lung-metastasis, which was verified by gene knockdown experiments in vivo. In addition, in vivo and in vitro functional analyses demonstrated that S100A14 enhanced scattering, motility and invasiveness of mouse tumor cells. Our model system may be adaptable to the diversity of metastasis in human cancers and useful for exploring the molecular mechanism responsible for metastasis.


Asunto(s)
Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Metástasis de la Neoplasia/genética , Proteínas S100/genética , Animales , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C3H , Invasividad Neoplásica
14.
Histol Histopathol ; 33(5): 497-505, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29181837

RESUMEN

Liver X receptors (LXRs) participate not only in maintaining cholesterol homeostasis but also in controlling cellular growth in many types of normal and tumor cells. We previously reported that LXRα was aberrantly expressed in human oral squamous cell carcinoma (HOSCC) tissues and cell lines, and that LXR stimulation led to significant reduction of proliferation of HOSCC cells via accelerating cholesterol efflux. Since LXRs and downstream proteins involved in cholesterol metabolism could be also applied as therapeutic targets in small cell lung carcinoma (SCLC) and pancreatic ductal adenocarcinoma (PDAC), we herein analyzed the distribution of LXR proteins in these refractory cancers as well as in normal human lung and pancreatic tissues. LXRß was observed in ciliated epithelial cells, bronchial gland epithelia, type II alveolar epithelia and alveolar macrophages of the lung, and was less expressed in bronchial basal cells and type I alveolar epithelia. In addition, LXRß was detected in epithelium of the pancreatic duct and acinar cells of the pancreas, and was weakly expressed in pancreatic islet cells. By contrast, LXRα expression was restricted to alveolar macrophages, and was not evident in any types of epithelial cells in the lung and pancreas. We also demonstrated that LXRß but not LXRα was abundantly expressed in nine cases of SCLC and twenty cases of PDAC tissues. These findings provide basic information for evaluating the efficacy of LXR-targeted treatment in SCLC and PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Carcinoma de Células Pequeñas/genética , Regulación Neoplásica de la Expresión Génica/genética , Receptores X del Hígado/biosíntesis , Receptores X del Hígado/genética , Neoplasias Pulmonares/genética , Neoplasias Pancreáticas/genética , Anciano , Anciano de 80 o más Años , Especificidad de Anticuerpos , ADN de Neoplasias/biosíntesis , ADN de Neoplasias/genética , Resistencia a Antineoplásicos/genética , Potenciales Postsinápticos Excitadores , Femenino , Humanos , Masculino , Persona de Mediana Edad
15.
Oncotarget ; 9(24): 16588-16598, 2018 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-29682171

RESUMEN

Since hepatitis C virus (HCV) is thought to enter into host hepatocytes using the same cellular pathways regardless of the genotypes, the host factors are promising targets to prevent and treat HCV infection. Human occludin (hOCLN) is one representative entry factor, and its second extracellular loop (EC2) contributes to the species selectivity of HCV-susceptibility. However, the exact function of hOCLN during HCV entry remains unknown, and no hOCLN-targeting antibodies or synthetic drugs that prevent and treat HCV infection have yet been developed. Here we generated the anti-hOCLN-EC2 monoclonal antibody (mAb) 67-2, and demonstrated that it efficiently inhibited HCV infection in the HCV-permissive human cell line Huh7.5.1. We also showed, using three different culture systems of Huh7.5.1 cells, that this novel mAb is accessible to OCLN from the basolateral side of hepatocytes but not from the apical side. In addition, our Western blot analyses indicated that the established 67-2 mAb reacted not only with hOCLN but also with mouse OCLN, strongly suggesting that 67-2 does not recognize the human-specific amino acids in OCLN-EC2. Moreover, we revealed that the anti-hOCLN-EC2 mAb 67-2 showed no adverse effects on cell viability or the barrier function of tight junctions.

16.
Oncotarget ; 8(55): 93382-93391, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29212157

RESUMEN

Schizophrenia is thought to be caused by a combination of genetic and environmental factors; however, its pathogenesis remains largely unknown. Here, we focus on the endothelial tight-junction protein claudin-5 (CLDN5), because the CLDN5 gene is mapped to the schizophrenia-associated 22q11.2 deletion region, and a single nucleotide polymorphism in the CLDN5 locus is also linked to schizophrenia. We show, by RT-qPCR and immunohistochemistry, that the expressions of CLDN5 mRNA and protein are significantly increased and decreased, respectively, in the schizophrenic prefrontal cortex (PFC) compared with control PFC. These changes were not observed in the schizophrenic visual cortex (VC), and neither the density nor diameter of the CD34-positive microvessels was altered in the schizophrenic PFC or VC. Interestingly, protein kinase A (PKA) was activated in the microvascular and perivascular regions of the schizophrenic PFC, and the pPKA-positive microvascular endothelial cells occasionally exhibited focal loss of CLND5. Since we previously demonstrated that cAMP induced CLDN5 mRNA expression and size-selective loosening of the endothelial barrier in PKA-independent and -dependent manners, respectively, a similar mechanism could contribute to the discrepancy between mRNA and protein expression of CLDN5 in the schizophrenic PFC. Taken collectively, these findings provide novel insights into the pathophysiology of schizophrenia.

17.
Oncotarget ; 6(32): 33345-57, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26452260

RESUMEN

Liver X receptors (LXRs) contribute not only to maintain cholesterol homeostasis but also to control cell growth. However, the molecular mechanisms behind the LXR-mediated anti-proliferative effects are largely unknown. Here we show, by immunohistochemistry, that LXRα and LXRß are differentially distributed in oral stratified squamous epithelia. By immunohistochemical and Western blot analyses, we also reveal that LXRα is abundantly expressed in human oral squamous cell carcinoma (HOSCC) tissues and cell lines. Cell counting, BrdU labeling and cell cycle assay indicated that LXR stimulation led to significant reduction of proliferation in HOSCC cells. Importantly, our study highlights, by using RNA interference, that the ATP-binding cassette transporter A1 (ABCA1)-accelerated cholesterol efflux is critical for the growth inhibitory action of LXRs in HOSCC cells. Moreover, we demonstrate that LXR activation reduces the growth of xenograft tumour of HOSCC cells in mice accompanied by the upregulation of ABCA1 expression and the decline of cholesterol levels in the tumour. These findings strongly suggested that targeting the LXR-regulated cholesterol transport, yielding in lowering intracellular cholesterol levels, could be a promising therapeutic option for certain types of cancers.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/genética , Carcinoma de Células Escamosas/patología , Proliferación Celular/genética , Colesterol/metabolismo , Neoplasias de la Boca/patología , Receptores Nucleares Huérfanos/fisiología , Transportador 1 de Casete de Unión a ATP/metabolismo , Adulto , Animales , Transporte Biológico/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Receptores X del Hígado , Masculino , Ratones , Ratones SCID , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Ratas , Ratas Wistar , Regulación hacia Arriba/genética
18.
PLoS One ; 8(10): e75106, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24116027

RESUMEN

During epithelialization, cell adhesions and polarity must be established to maintain tissue assemblies and separate the biological compartments in the body. However, the molecular basis of epithelial morphogenesis, in particular, a role of cell adhesion molecules in epithelial differentiation from stem cells, remains unclear. Here, we show that the stable and conditional expression of a tight-junction protein, claudin-6 (Cldn6), triggers epithelial morphogenesis in mouse F9 stem cells. We also demonstrate that Cldn6 induces the expression of other tight-junction and microvillus molecules including Cldn7, occludin, ZO-1α+, and ezrin/radixin/moesin-binding phosphoprotein50. These events were inhibited by attenuation of Cldn6 using RNA interference or the C-terminal half of Clostridium Perfringens enterotoxin. Furthermore, similar results were obtained in mouse embryonic stem cells. Thus, we have uncovered that the Cldn6 functions as a novel cue to induce epithelial differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Claudinas/metabolismo , Células Madre Embrionarias/citología , Células Epiteliales/citología , Animales , Claudinas/genética , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Células Madre Embrionarias/metabolismo , Células Epiteliales/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ocludina/genética , Ocludina/metabolismo , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
19.
PLoS One ; 7(9): e45517, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029067

RESUMEN

The human synovium contains mesenchymal stem cells (MSCs), which are multipotential non-hematopoietic progenitor cells that can differentiate into a variety of mesenchymal lineages and they may therefore be a candidate cell source for tissue repair. However, the molecular mechanisms by which this can occur are still largely unknown. Mouse primary cell culture enables us to investigate the molecular mechanisms underlying various phenomena because it allows for relatively easy gene manipulation, which is indispensable for the molecular analysis. However, mouse synovial mesenchymal cells (SMCs) have not been established, although rabbit, cow, and rat SMCs are available, in addition to human MSCs. The aim of this study was to establish methods to harvest the synovium and to isolate and culture primary SMCs from mice. As the mouse SMCs were not able to be harvested and isolated using the same protocol for human, rat and rabbit SMCs, the protocol for humans was modified for SMCs from the Balb/c mouse knee joint. The mouse SMCs obtained showed superior proliferative potential, growth kinetics and colony formation compared to cells derived from muscle and bone marrow. They expressed PDGFRá and Sca-1 detected by flow cytometry, and showed an osteogenic, adipogenic and chondrogenic potential similar or superior to the cells derived from muscle and bone marrow by demonstrating in vitro osteogenesis, adipogenesis and chondrogenesis. In conclusion, we established a primary mouse synovial cell culture method. The cells derived from the mouse synovium demonstrated both the ability to proliferate and multipotentiality similar or superior to the cells derived from muscle and bone marrow.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Membrana Sinovial/citología , Adipogénesis/fisiología , Animales , Diferenciación Celular/fisiología , Separación Celular/métodos , Condrogénesis/fisiología , Femenino , Inmunofenotipificación , Articulación de la Rodilla/anatomía & histología , Articulación de la Rodilla/citología , Ratones , Ratones Endogámicos BALB C , Osteogénesis/fisiología , Cultivo Primario de Células
20.
Matrix Biol ; 31(1): 17-28, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21983115

RESUMEN

Laminin α1 (Lama1), which is a subunit of laminin-1 (laminin-111), a heterotrimeric ECM protein, is essential for embryonic development and promotes neurite outgrowth in culture. Because the deletion of Lama1 causes lethality at early embryonic stages in mice, the in vivo role of Lama1 in neural development and functions has not yet been possible to determine. In this study, we generated conditional Lama1 knockout (Lama1(CKO)) mice in the epiblast lineage using Sox2-Cre mice. These Lama1(CKO) mice survived, but displayed behavioral disorders and impaired formation of the cerebellum. Deficiency of Lama1 in the pial basement membrane of the meninges resulted in defects in the conformation of the meninges. During cerebellar development, Lama1 deficiency also caused a decrease in the proliferation and migration of granule cell precursors, disorganization of Bergmann glial fibers and endfeet, and a transient reduction in the activity of Akt. A marked reduction in numbers of dendritic processes in Purkinje cells was observed in Lama1(CKO) mice. Together, these results indicate that Lama1 is required for cerebellar development and functions.


Asunto(s)
Cerebelo/anomalías , Cerebelo/crecimiento & desarrollo , Laminina/metabolismo , Meninges/anomalías , Meninges/crecimiento & desarrollo , Animales , Membrana Basal/metabolismo , Conexinas/metabolismo , Dendritas/metabolismo , Dendritas/patología , Laminina/deficiencia , Laminina/genética , Ratones , Ratones Noqueados/genética , Neuritas/patología , Proteína Oncogénica v-akt/metabolismo , Células de Purkinje/metabolismo , Células de Purkinje/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA