Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Molecules ; 29(2)2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-38257315

RESUMEN

Collagen is an important material for biomedical research, but using mammalian tissue-derived collagen carries the risk of zoonotic disease transmission. Marine organisms, such as farmed tilapia, have emerged as a safe alternative source of collagen for biomedical research. However, the tilapia collagen products for biomedical research are rare, and their biological functions remain largely unexamined. In this study, we characterized a commercial tilapia skin collagen using SDS-PAGE and fibril formation assays and evaluated its effects on skin fibroblast adhesion, proliferation, and migration, comparing it with commercial collagen from rat tails, porcine skin, and bovine skin. The results showed that tilapia skin collagen is a type I collagen, similar to rat tail collagen, and has a faster fibril formation rate and better-promoting effects on cell migration than porcine and bovine skin collagen. We also confirmed its application in a 3D culture for kidney cells' spherical cyst formation, fibroblast-induced gel contraction, and tumor spheroid interfacial invasion. Furthermore, we demonstrated that the freeze-dried tilapia skin collagen scaffold improved wound closure in a mouse excisional wound model, similar to commercial porcine or bovine collagen wound dressings. In conclusion, tilapia skin collagen is an ideal biomaterial for biomedical research.


Asunto(s)
Investigación Biomédica , Tilapia , Ratones , Ratas , Porcinos , Animales , Bovinos , Mamíferos , Colágeno/farmacología , Piel , Modelos Animales de Enfermedad
2.
Biomedicines ; 10(5)2022 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-35625714

RESUMEN

We demonstrate that Ha-RasV12 overexpression induces the nuclear translocation of Hippo effector Yes-associated protein (YAP) in MDCK cells via the hippo-independent pathway at the confluent stage. Ha-RasV12 overexpression leads to the downregulation of Caveolin-1 (Cav1) and the disruption of junction integrity. It has been shown that the disruption of actin belt integrity causes YAP nuclear translocation in epithelial cells at high density. Therefore, we hypothesized that Ha-RasV12-decreased Cav1 leads to the disruption of cell junction integrity, which subsequently facilitates YAP nuclear retention. We revealed that Ha-RasV12 downregulated Cav1 through the ERK pathway. Furthermore, the distribution and expression of Cav1 mediated the cell junction integrity and YAP nuclear localization. This suggests that the downregulation of Cav1 induced by Ha-RasV12 disrupted the cell junction integrity and promoted YAP nuclear translocation. We further indicated the consequence of Ha-RasV12-induced YAP activation. Surprisingly, the activation of YAP is not required for Ha-RasV12-induced multilayer cellular aggregates. Instead, Ha-RasV12 triggered the ERK-Rac pathway to promote cellular aggregate formation. Moreover, the overexpression of constitutively active Rac is sufficient to trigger cellular aggregation in MDCK cells at the confluent stage. This highlights that Rac activity is essential for cellular aggregates.

4.
Biochim Biophys Acta Mol Cell Res ; 1866(11): 118473, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30954568

RESUMEN

Discoidin domain receptors DDR1 and DDR2 are collagen receptor tyrosine kinases that have many roles in tissue development and disease progression. Under physiological conditions, DDR1 is predominantly expressed in epithelial cells and functions to maintain cell differentiation and tissue homeostasis. A switch in expression from DDR1 to DDR2 occurs during epithelial-to-mesenchymal transition. However, opposite effects of DDR1 are reported to be involved in the progression of cancer and fibrotic diseases. Accumulating evidence suggests that DDR1 is involved in pro-metastasis and pro-survival signals. This review summarizes the roles of DDR1 in epithelial cell differentiation, cell migration, cancer progression and tissues fibrosis and highlights how the dichotomous functions of DDR1 may relevant to different cell types and statues. Elucidation of the underlying mechanism of the dichotomous functions of DDR1 will help to develop DDR1 as a therapeutic target.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Receptor con Dominio Discoidina 1/fisiología , Progresión de la Enfermedad , Fibrosis/metabolismo , Neoplasias/metabolismo , Adhesión Celular , Colágeno , Receptor con Dominio Discoidina 2 , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Integrina beta1 , Metástasis de la Neoplasia , Proteínas Tirosina Quinasas Receptoras
5.
Cell Death Discov ; 4: 37, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29531834

RESUMEN

Ca2+ plays a significant role in linking the induction of apoptosis. The key anti-apoptotic protein, Bcl-2, has been reported to regulate the movement of Ca2+ across the ER membrane, but the exact effect of Bcl-2 on Ca2+ levels remains controversial. Store-operated Ca2+ entry (SOCE), a major mode of Ca2+ uptake in non-excitable cells, is activated by depletion of Ca2+ in the ER. Depletion of Ca2+ in the ER causes translocation of the SOC channel activator, STIM1, to the plasma membrane. Thereafter, STIM1 binds to Orai1 or/and TRPC1 channels, forcing them to open and thereby allow Ca2+ entry. In addition, several anti-cancer drugs have been reported to induce apoptosis of cancer cells via the SOCE pathway. However, the detailed mechanism underlying the regulation of SOCE by Bcl-2 is not well understood. In this study, a three-amino acid mutation within the Bcl-2 BH1 domain was generated to verify the role of Bcl-2 in Ca2+ handling during ER stress. The subcellular localization of the Bcl-2 mutant (mt) is similar to that in the wild-type Bcl-2 (WT) in the ER and mitochondria. We found that mt enhanced thapsigargin and tunicamycin-induced apoptosis through ER stress-mediated apoptosis but not through the death receptor- and mitochondria-dependent apoptosis, while WT prevented thapsigargin- and tunicamycin-induced apoptosis. In addition, mt depleted Ca2+ in the ER lumen and also increased the expression of SOCE-related molecules. Therefore, a massive Ca2+ influx via SOCE contributed to caspase activation and apoptosis. Furthermore, inhibiting SOCE or chelating either extracellular or intracellular Ca2+ inhibited mt-mediated apoptosis. In brief, our results explored the critical role of Bcl-2 in Ca2+ homeostasis and the modulation of ER stress.

6.
J Dermatol Sci ; 90(3): 232-240, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29567352

RESUMEN

Mechanical forces are known to regulate homeostasis of the skin and play a role in the pathogenesis of skin diseases. The epidermis consists of keratinocytes that are tightly adhered to each other by cell junctions. Defects in keratins or desmosomal/hemidesmosomal proteins lead to the attenuation of mechanical strength and formation of intraepidermal blisters in the case of epidermolysis bullosa simplex. The dermis is rich in extracellular matrix, especially collagen, and provides the majority of tensile force in the skin. Keloid and hypertrophic scar, which is the result of over-production of collagen by fibroblasts during the wound healing, are associated with extrinsic tensile forces and changes of intrinsic mechanical properties of the cell. Increasing evidences shows that stiffness of the skin environment determines the regenerative ability during wound healing process. Mechanotransduction pathways are also involved in the morphogenesis and cyclic growth of hair follicles. The development of androgenetic alopecia is correlated to tensile forces generated by the fibrous tissue underlying the scalp. Acral melanoma predominantly occurs in the weight-bearing area of the foot suggesting the role of mechanical stress. Increased dermal stiffness from fibrosis might be the cause of recessive dystrophic epidermolysis bullosa associated squamous cell carcinoma. Strategies to change the mechanical forces or modify the mechanotransduction signals may lead to a new way to treat skin diseases and promote skin regeneration.


Asunto(s)
Desmosomas/patología , Mecanotransducción Celular , Enfermedades de la Piel/patología , Piel/patología , Cicatrización de Heridas/fisiología , Colágeno/metabolismo , Matriz Extracelular , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Queratinocitos/patología , Queratinas/metabolismo , Piel/citología
7.
J Cell Mol Med ; 22(5): 2631-2643, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29502342

RESUMEN

Caveolin-1 (Cav1) is down-regulated during MK4 (MDCK cells harbouring inducible Ha-RasV12 gene) transformation by Ha-RasV12 . Cav1 overexpression abrogates the Ha-RasV12 -driven transformation of MK4 cells; however, the targeted down-regulation of Cav1 is not sufficient to mimic this transformation. Cav1-silenced cells, including MK4/shCav1 cells and MDCK/shCav1 cells, showed an increased cell area and discontinuous junction-related proteins staining. Cellular and mechanical transformations were completed when MDCK/shCav1 cells were treated with medium conditioned by MK4 cells treated with IPTG (MK4+I-CM) but not with medium conditioned by MK4 cells. Nanoparticle tracking analysis showed that Ha-RasV12 -inducing MK4 cells increased exosome-like microvesicles release compared with their normal counterparts. The cellular and mechanical transformation activities of MK4+I-CM were abolished after heat treatment and exosome depletion and were copied by exosomes derived from MK4+I-CM (MK4+I-EXs). Wnt5a, a downstream product of Ha-RasV12 , was markedly secreted by MK4+I-CM and MK4+I-EXs. Suppression of Wnt5a expression and secretion using the porcupine inhibitor C59 or Wnt5a siRNA inhibited the Ha-RasV12 - and MK4+I-CM-induced transformation of MK4 cells and MDCK/shCav1 cells, respectively. Cav1 down-regulation, either by Ha-RasV12 or targeted shRNA, increased frizzled-2 (Fzd2) protein levels without affecting its mRNA levels, suggesting a novel role of Cav1 in negatively regulating Fzd2 expression. Additionally, silencing Cav1 facilitated the internalization of MK4+I-EXs in MDCK cells. These data suggest that Cav1-dependent repression of Fzd2 and exosome uptake is potentially relevant to its antitransformation activity, which hinders the activation of Ha-RasV12 -Wnt5a-Stat3 pathway. Altogether, these results suggest that both decreasing Cav1 and increasing exosomal Wnt5a must be implemented during Ha-RasV12 -driven cell transformation.


Asunto(s)
Caveolina 1/genética , Transformación Celular Neoplásica/genética , Regulación hacia Abajo/genética , Receptores Frizzled/metabolismo , Transducción de Señal , Proteína Wnt-5a/metabolismo , Proteínas ras/metabolismo , Animales , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Medios de Cultivo Condicionados/farmacología , Perros , Regulación hacia Abajo/efectos de los fármacos , Exosomas/efectos de los fármacos , Exosomas/metabolismo , Humanos , Isopropil Tiogalactósido/farmacología , Células de Riñón Canino Madin Darby , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba/efectos de los fármacos
8.
J Invest Dermatol ; 138(1): 208-218, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28899682

RESUMEN

Keloids are pathological scars characterized by excessive extracellular matrix production that are prone to form in body sites with increased skin tension. CAV1, the principal coat protein of caveolae, has been associated with the regulation of cell mechanics, including cell softening and loss of stiffness sensing ability in NIH3T3 fibroblasts. Although CAV1 is present in low amounts in keloid fibroblasts (KFs), the causal association between CAV1 down-regulation and its aberrant responses to mechanical stimuli remain unclear. In this study, atomic force microscopy showed that KFs were softer than normal fibroblasts with a loss of stiffness sensing. The decrease of CAV1 contributed to the hyperactivation of fibrogenesis-associated RUNX2, a transcription factor germane to osteogenesis/chondrogenesis, and increased migratory ability in KFs. Treatment of KFs with trichostatin A, which increased the acetylation level of histone H3, increased CAV1 and decreased RUNX2 and fibronectin. Trichostatin A treatment also resulted in cell stiffening and decreased migratory ability in KFs. Collectively, these results suggest a role for CAV1 down-regulation in linking the aberrant responsiveness to mechanical stimulation and extracellular matrix accumulation with the progression of keloids, findings that may lead to new developments in the prevention and treatment of keloid scarring.


Asunto(s)
Caveolina 1/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Fibroblastos/patología , Queloide/patología , Adulto , Biopsia , Caveolina 1/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Regulación hacia Abajo , Femenino , Fibroblastos/ultraestructura , Técnicas de Silenciamiento del Gen , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Microscopía de Fuerza Atómica , Persona de Mediana Edad , Cultivo Primario de Células , ARN Interferente Pequeño/metabolismo , Piel/citología , Piel/patología , Adulto Joven
9.
Sci Rep ; 7(1): 15008, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118431

RESUMEN

Focal adhesion (FA) assembly, mediated by integrin activation, responds to matrix stiffness; however, the underlying mechanisms are unclear. Here, we showed that ß1 integrin and caveolin-1 (Cav1) levels were decreased with declining matrix stiffness. Soft matrix selectively downregulated ß1 integrin by endocytosis and subsequent lysosomal degradation. Disruption of lipid rafts with methyl-ß-cyclodextrin or nystatin, or knockdown of Cav1 by siRNA decreased cell spreading, FA assembly, and ß1 integrin protein levels in cells cultured on stiff matrix. Overexpression of Cav1, particularly the phospho-mimetic mutant Cav1-Y14D, averted soft matrix-induced decreases in ß1 integrin protein levels, cell spreading, and FA assembly in NMuMG cells. Interestingly, overexpression of an auto-clustering ß1 integrin hindered soft matrix-induced reduction of Cav1 and cell spreading, which suggests a reciprocal regulation between ß1 integrin and Cav1. Finally, co-expression of this auto-clustering ß1 integrin and Cav1-Y14D synergistically enhanced cell spreading, and FA assembly in HEK293T cells cultured on either stiff ( > G Pa) or soft (0.2 kPa) matrices. Collectively, these results suggest that matrix stiffness governs the expression of ß1 integrin and Cav1, which reciprocally control each other, and subsequently determine FA assembly and turnover.


Asunto(s)
Caveolina 1/metabolismo , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Integrina beta1/metabolismo , Mecanotransducción Celular , Animales , Caveolina 1/genética , Línea Celular , Movimiento Celular/genética , Endocitosis/genética , Matriz Extracelular/química , Células HEK293 , Humanos , Integrina beta1/genética , Microdominios de Membrana/metabolismo , Ratones , Mutación , Interferencia de ARN
10.
Kidney Int ; 91(2): 412-422, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28341240

RESUMEN

Neutrophil gelatinase-associated lipocalin (Ngal) is a biomarker for acute and chronic renal injuries, including polycystic kidney disease (PKD). However, the effect of Ngal on PKD progression remains unexplored. To study this, we generated 3 strains of mice with different expression levels of Ngal within an established PKD model (Pkd1L3/L3): Pkd1L3/L3 (with endogenous Ngal), Pkd1L3/L3; NgalTg/Tg (with endogenous and overexpression of exogenous kidney-specific Ngal) and Pkd1L3/L3; Ngal-/- mice (with Ngal deficiency). Knockout of endogenous Ngal had no effect on phenotypes, cystic progression, or survival of the PKD mice. However, the transgenic mice had a significantly longer lifespan, smaller (but not fewer) renal cysts, and less interstitial fibrosis than the mice without or with endogenous Ngal. Western-blot analyses showed significant increases in Ngal and cleaved caspase-3 and decreases in α-smooth muscle actin, hypoxia-inducible factor 1-α, pro-caspase 3, proliferating cell nuclear antigen, Akt, mammalian target of rapamycin, and S6 Kinase in the transgenic mice as compared with the other 2 strains of PKD mice. Thus, overexpression of exogenous kidney-specific Ngal reduced cystic progression and prolonged the lifespan in PKD mice, was associated with reductions in interstitial fibrosis and proliferation, and augmented apoptosis.


Asunto(s)
Riñón/metabolismo , Lipocalina 2/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Actinas/metabolismo , Animales , Apoptosis , Cadherinas/genética , Caspasa 3/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Femenino , Fibrosis , Predisposición Genética a la Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Riñón/patología , Lipocalina 2/genética , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Fosforilación , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Regiones Promotoras Genéticas , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Factores de Tiempo
11.
Cell Adh Migr ; 10(4): 368-77, 2016 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-26919488

RESUMEN

Any cellular response leading to morphological changes is highly tuned to balance the force generated from structural reorganization, provided by actin cytoskeleton. Actin filaments serve as the backbone of intracellular force, and transduce external mechanical signal via focal adhesion complex into the cell. During migration, cells not only undergo molecular changes but also rapid mechanical modulation. Here we focus on determining, the role of spatial distribution of mechanical changes of actin filaments in epithelial, mesenchymal, fibrotic and cancer cells with non-migration, directional migration, and non-directional migration behaviors using the atomic force microscopy. We found 1) non-migratory cells only generated one type of filament elasticity, 2) cells generating spatially distributed two types of filament elasticity showed directional migration, and 3) pathologic cells that autonomously generated two types of filament elasticity without spatial distribution were actively migrating non-directionally. The demonstration of spatial regulation of filament elasticity of different cell types at the nano-scale highlights the coupling of cytoskeletal function with physical characters at the sub-cellular level, and provides new research directions for migration related disease.


Asunto(s)
Movimiento Celular , Elasticidad , Actinas/metabolismo , Animales , Línea Celular , Polaridad Celular , Fibroblastos/patología , Humanos , Queloide/patología , Ratones , Microtúbulos/metabolismo , Osteosarcoma/patología , Polimerizacion
12.
Am J Pathol ; 185(9): 2468-81, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26212907

RESUMEN

The proliferation of mouse proximal tubular epithelial cells in ex vivo culture depends on matrix stiffness. Combined analysis of the microarray and experimental data revealed that Krüppel-like factor (Klf)5 was the most up-regulated transcription factor accompanied by the down-regulation of Klf4 when cells were on stiff matrix. These changes were reversed by soft matrix via extracellular signal-regulated kinase (ERK) inactivation. Knockdown of Klf5 or forced expression of Klf4 inhibited stiff matrix-induced cell spreading and proliferation, suggesting that Klf5/Klf4 act as positive and negative regulators, respectively. Moreover, stiff matrix-activated ERK increased the protein level and nuclear translocation of mechanosensitive Yes-associated protein 1 (YAP1), which is reported to prevent Klf5 degradation. Finally, in vivo model of unilateral ureteral obstruction revealed that matrix stiffness-regulated Klf5/Klf4 is related to the pathogenesis of renal fibrosis. In the dilated tubules of obstructed kidney, ERK/YAP1/Klf5/cyclin D1 axis was up-regulated and Klf4 was down-regulated. Inhibition of collagen crosslinking by lysyl oxidase inhibitor alleviated unilateral ureteral obstruction-induced tubular dilatation and proliferation, preserved Klf4, and suppressed the ERK/YAP1/Klf5/cyclin D1 axis. This study unravels a novel mechanism how matrix stiffness regulates cellular proliferation and highlights the importance of matrix stiffness-modulated Klf5/Klf4 in the regulation of renal physiologic functions and fibrosis progression.


Asunto(s)
Enfermedades Renales/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Proliferación Celular/fisiología , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis/metabolismo , Enfermedades Renales/patología , Factor 4 Similar a Kruppel , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Regulación hacia Arriba
13.
Oncotarget ; 6(25): 20946-58, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26189182

RESUMEN

The stiffness sensing ability is required to respond to the stiffness of the matrix. Here we determined whether normal cells and cancer cells display distinct mechanical phenotypes. Cancer cells were softer than their normal counterparts, regardless of the type of cancer (breast, bladder, cervix, pancreas, or Ha-RasV12-transformed cells). When cultured on matrices of varying stiffness, low stiffness decreased proliferation in normal cells, while cancer cells and transformed cells lost this response. Thus, cancer cells undergo a change in their mechanical phenotype that includes cell softening and loss of stiffness sensing. Caveolin-1, which is suppressed in many tumor cells and in oncogene-transformed cells, regulates the mechanical phenotype. Caveolin-1-upregulated RhoA activity and Y397FAK phosphorylation directed actin cap formation, which was positively correlated with cell elasticity and stiffness sensing in fibroblasts. Ha-RasV12-induced transformation and changes in the mechanical phenotypes were reversed by re-expression of caveolin-1 and mimicked by the suppression of caveolin-1 in normal fibroblasts. This is the first study to describe this novel role for caveolin-1, linking mechanical phenotype to cell transformation. Furthermore, mechanical characteristics may serve as biomarkers for cell transformation.


Asunto(s)
Caveolina 1/metabolismo , Neoplasias/patología , Proteínas ras/metabolismo , Actinas/química , Animales , Fenómenos Biomecánicos , Proliferación Celular , Transformación Celular Neoplásica , Colágeno/química , Perros , Elasticidad , Inhibidores Enzimáticos/química , Femenino , Fibroblastos/metabolismo , Genes ras , Humanos , Células de Riñón Canino Madin Darby , Ratones , Microscopía de Fuerza Atómica , Microscopía Confocal , Microscopía Fluorescente , Células 3T3 NIH , Neoplasias/metabolismo , Fenotipo , Fosforilación , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Proteína de Unión al GTP rhoA/metabolismo
14.
Oncotarget ; 6(18): 15966-83, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-25965826

RESUMEN

Modulations of cytoskeletal organization and focal adhesion turnover correlate to tumorigenesis and epithelial-mesenchymal transition (EMT), the latter process accompanied by the loss of epithelial markers and the gain of mesenchymal markers (e.g., vimentin). Clinical microarray results demonstrated that increased levels of vimentin mRNA after chemotherapy correlated to a poor prognosis of breast cancer patients. We hypothesized that vimentin mediated the reorganization of cytoskeletons to maintain the mechanical integrity in EMT cancer cells. By using knockdown strategy, the results showed reduced cell proliferation, impaired wound healing, loss of directional migration, and increased large membrane extension in MDA-MB 231 cells. Vimentin depletion also induced reorganization of cytoskeletons and reduced focal adhesions, which resulted in impaired mechanical strength because of reduced cell stiffness and contractile force. In addition, overexpressing vimentin in MCF7 cells increased cell stiffness, elevated cell motility and directional migration, reoriented microtubule polarity, and increased EMT phenotypes due to the increased ß1-integrin and the loss of junction protein E-cadherin. The EMT-related transcription factor slug was also mediated by vimentin. The current study demonstrated that vimentin serves as a regulator to maintain intracellular mechanical homeostasis by mediating cytoskeleton architecture and the balance of cell force generation in EMT cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Citoesqueleto/metabolismo , Adhesiones Focales/fisiología , Vimentina/metabolismo , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Citoesqueleto/patología , Transición Epitelial-Mesenquimal/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Microtúbulos/metabolismo , Microtúbulos/patología , Vimentina/deficiencia , Vimentina/genética
15.
Exp Dermatol ; 24(8): 579-84, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25877039

RESUMEN

One of the key features of keloid is its fibroblasts migrating beyond the original wound border. During migration, cells not only undergo molecular changes but also mechanical modulation. This process is led by actin filaments serving as the backbone of intra-cellular force and transduces external mechanical signal via focal adhesion complex into the cell. Here, we focus on determining the mechanical changes of actin filaments and the spatial distribution of forces in response to changing chemical stimulations and during cell migration. Atomic force microscopy and micropost array detector are used to determine and compare the magnitude and distribution of filament elasticity and force generation in fibroblasts and keloid fibroblasts. We found both filament elasticity and force generation show spatial distribution in a polarized and migrating cell. Such spatial distribution is disrupted when mechano-signalling is perturbed by focal adhesion kinase inhibitor and in keloid fibroblasts. The demonstration of keloid pathology at the nanoscale highlights the coupling of cytoskeletal function with physical characters at the subcellular level and provides new research directions for migration-related disease such as keloid.


Asunto(s)
Citoesqueleto/fisiología , Fibroblastos/fisiología , Queloide/patología , Citoesqueleto de Actina/fisiología , Animales , Movimiento Celular , Polaridad Celular , Elasticidad , Fibroblastos/ultraestructura , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/fisiología , Adhesiones Focales/fisiología , Humanos , Ratones , Microscopía de Fuerza Atómica , Células 3T3 NIH , Quinolonas/farmacología , Estrés Mecánico , Sulfonas/farmacología , Cicatrización de Heridas
16.
J Cell Physiol ; 230(9): 2086-97, 2015 09.
Artículo en Inglés | MEDLINE | ID: mdl-25639747

RESUMEN

Ca(2+) -mediated formation of cell polarity is essential for directional migration which plays an important role in physiological and pathological processes in organisms. To examine the critical role of store-operated Ca(2+) entry, which is the major form of extracellular Ca(2+) influx in non-excitable cells, in the formation of cell polarity, we employed human bone osteosarcoma U2OS cells, which exhibit distinct morphological polarity during directional migration. Our analyses showed that Ca(2+) was concentrated at the rear end of cells and that extracellular Ca(2+) influx was important for cell polarization. Inhibition of store-operated Ca(2+) entry using specific inhibitors disrupted the formation of cell polarity in a dose-dependent manner. Moreover, the channelosomal components caveolin-1, TRPC1, and Orai1 were concentrated at the rear end of polarized cells. Knockdown of TRPC1 or a TRPC inhibitor, but not knockdown of Orai1, reduced cell polarization. Furthermore, disruption of lipid rafts or overexpression of caveolin-1 contributed to the downregulation of cell polarity. On the other hand, we also found that cell polarity, store-operated Ca(2+) entry activity, and cell stiffness were markedly decreased by low substrate rigidity, which may be caused by the disorganization of actin filaments and microtubules that occurs while regulating the activity of the mechanosensitive TRPC1 channel.


Asunto(s)
Calcio/metabolismo , Polaridad Celular/genética , Mecanotransducción Celular/genética , Osteosarcoma/genética , Canales de Calcio/genética , Señalización del Calcio/genética , Caveolina 1/genética , Línea Celular Tumoral , Humanos , Proteína ORAI1 , Osteosarcoma/patología , ARN Interferente Pequeño , Canales Catiónicos TRPC/genética
17.
Am J Physiol Renal Physiol ; 307(6): F695-707, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25056346

RESUMEN

To explore whether matrix stiffness affects cell differentiation, proliferation, and transforming growth factor (TGF)-ß1-induced epithelial-mesenchymal transition (EMT) in primary cultures of mouse proximal tubular epithelial cells (mPTECs), we used a soft matrix made from monomeric collagen type I-coated polyacrylamide gel or matrigel (MG). Both kinds of soft matrix benefited primary mPTECs to retain tubular-like morphology with differentiation and growth arrest and to evade TGF-ß1-induced EMT. However, the potent effect of MG on mPTEC differentiation was suppressed by glutaraldehyde-induced cross-linking and subsequently stiffening MG or by an increasing ratio of collagen in the soft mixed gel. Culture media supplemented with MG also helped mPTECs to retain tubular-like morphology and a differentiated phenotype on stiff culture dishes as soft MG did. We further found that the protein level and activity of ERK were scaled with the matrix stiffness. U-0126, a MEK inhibitor, abolished the stiff matrix-induced dedifferentiation and proliferation. These data suggest that the ERK signaling pathway plays a vital role in matrix stiffness-regulated cell growth and differentiation. Taken together, both compliant property and specific MG signals from the matrix are required for the regulation of epithelial differentiation and proliferation. This study provides a basic understanding of how physical and chemical cues derived from the extracellular matrix regulate the physiological function of proximal tubules and the pathological development of renal fibrosis.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Matriz Extracelular/fisiología , Túbulos Renales Proximales/citología , Animales , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Porcinos , Factor de Crecimiento Transformador beta1
18.
PLoS One ; 8(10): e77384, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24194882

RESUMEN

Atomic force microscopy provides a novel technique for differentiating the mechanical properties of various cell types. Cell elasticity is abundantly used to represent the structural strength of cells in different conditions. In this study, we are interested in whether physical or physiological cues affect cell elasticity in Atomic force microscopy (AFM)-based assessments. The physical cues include the geometry of the AFM tips, the indenting force and the operating temperature of the AFM. All of these cues show a significant influence on the cell elasticity assessment. Sharp AFM tips create a two-fold increase in the value of the effective Young's modulus (E(eff)) relative to that of the blunt tips. Higher indenting force at the same loading rate generates higher estimated cell elasticity. Increasing the operation temperature of the AFM leads to decreases in the cell stiffness because the structure of actin filaments becomes disorganized. The physiological cues include the presence of fetal bovine serum or extracellular matrix-coated surfaces, the culture passage number, and the culture density. Both fetal bovine serum and the extracellular matrix are critical for cells to maintain the integrity of actin filaments and consequently exhibit higher elasticity. Unlike primary cells, mouse kidney progenitor cells can be passaged and maintain their morphology and elasticity for a very long period without a senescence phenotype. Finally, cell elasticity increases with increasing culture density only in MDCK epithelial cells. In summary, for researchers who use AFM to assess cell elasticity, our results provide basic and significant information about the suitable selection of physical and physiological cues.


Asunto(s)
Elasticidad/fisiología , Riñón/citología , Microscopía de Fuerza Atómica/métodos , Células Madre/citología , Citoesqueleto de Actina/fisiología , Animales , Fenómenos Biomecánicos , Bovinos , Recuento de Células , Perros , Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Células de Riñón Canino Madin Darby , Ratones
19.
Am J Physiol Cell Physiol ; 303(12): C1207-17, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23015544

RESUMEN

As increase in collagen deposition is no longer taken as simply a consequence but, rather, an inducer of disease progression; therefore, the understanding of collagen signal transduction is fundamentally important. Cells contain at least two types of collagen receptors: integrins and discoidin domain receptors (DDRs). The integrin heterodimers α(1)ß(1), α(2)ß(1), α(10)ß(1), and α(11)ß(1) are recognized as the non-tyrosine kinase collagen receptors. DDR1 and 2, the tyrosine kinase receptors of collagen, are specifically expressed in epithelium and mesenchyme, respectively. While integrin ß(1) and DDR1 are both required for cell adhesion on collagen, their roles in epithelial cell differentiation during development and disease progression seem to counteract each other, with integrin ß(1) favoring epithelium mesenchyme transition (EMT) and DDR1 inducing epithelial cell differentiation. The in vitro evidence shows that the integrin ß(1) and DDR1 exert opposing actions in regulation of membrane stability of E-cadherin, which itself is a critical regulator of epithelial cell differentiation. Here, we review the functional roles of integrin ß(1) and DDR1 in regulation of epithelial cell differentiation during development and disease progression, and explore the underlining mechanisms regarding to the regulation of membrane stability of E-cadherin.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/fisiología , Integrina beta1/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Colágeno/fisiología , Receptores Mitogénicos/fisiología , Animales , Cadherinas/fisiología , Colágeno/fisiología , Receptores con Dominio Discoidina , Femenino , Fibrosis , Humanos , Mesodermo/fisiología , Ratones , Neoplasias/patología
20.
PLoS One ; 7(3): e34011, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22479503

RESUMEN

It is estimated that 10-15% of couples are infertile and male factors account for about half of these cases. With the advent of intracytoplasmic sperm injection (ICSI), many infertile men have been able to father offspring. However, teratozoospermia still remains a big challenge to tackle. Septins belong to a family of cytoskeletal proteins with GTPase activity and are involved in various biological processes e.g. morphogenesis, compartmentalization, apoptosis and cytokinesis. SEPTIN12, identified by c-DNA microarray analysis of infertile men, is exclusively expressed in the post meiotic male germ cells. Septin12(+/+)/Septin12(+/-) chimeric mice have multiple reproductive defects including the presence of immature sperm in the semen, and sperm with bent neck (defect of the annulus) and nuclear DNA damage. These facts make SEPTIN12 a potential sterile gene in humans. In this study, we sequenced the entire coding region of SEPTIN12 in infertile men (n = 160) and fertile controls (n = 200) and identified ten variants. Among them is the c.474 G>A variant within exon 5 that encodes part of the GTP binding domain. The variant creates a novel splice donor site that causes skipping of a portion of exon 5, resulting in a truncated protein lacking the C-terminal half of SEPTIN12. Most individuals homozygous for the c.474 A allele had teratozoospermia (abnormal sperm <14%) and their sperm showed bent tail and de-condensed nucleus with significant DNA damage. Ex vivo experiment showed truncated SEPT12 inhibits filament formation in a dose-dependent manner. This study provides the first causal link between SEPTIN12 genetic variant and male infertility with distinctive sperm pathology. Our finding also suggests vital roles of SEPT12 in sperm nuclear integrity and tail development.


Asunto(s)
Astenozoospermia/genética , Predisposición Genética a la Enfermedad , Variación Genética , Infertilidad Masculina/genética , Septinas/genética , Espermatozoides/anomalías , Alelos , Apoptosis , Estudios de Casos y Controles , Núcleo Celular/metabolismo , Citocinesis , Citoesqueleto/metabolismo , Daño del ADN , ADN Complementario/metabolismo , GTP Fosfohidrolasas/metabolismo , Homocigoto , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Semen/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA