Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 81(22): 5666-5677, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34385183

RESUMEN

Collagen remodeling contributes to many physiologic and pathologic processes. In primary tumors, the linearization of collagen fibers promotes cancer cell invasion and metastasis and is indicative of poor prognosis. However, it remains unknown whether there are endogenous inhibitors of collagen linearization that could be exploited therapeutically. Here, we show that collagen linearization is controlled by two secreted matricellular proteins with antagonistic functions. Specifically, WISP1 was secreted by cancer cells, bound to type I collagen (Col I), and linearized Col I via its cysteine-rich C-terminal (CT) domain. In contrast, WISP2, which lacks a CT domain, inhibited Col I linearization by preventing WISP1-Col I binding. Analysis of patient data revealed that WISP2 expression is lower in most solid tumors, in comparison with normal tissues. Consequently, genetic or pharmacologic restoration of higher WISP2 levels impaired collagen linearization and prevented tumor cell invasion and metastasis in vivo in models of human and murine breast cancer. Thus, this study uncovers WISP2 as the first inhibitor of collagen linearization ever identified and reveals that collagen architecture can be normalized and metastasis inhibited by therapeutically restoring a high WISP2:WISP1 ratio. SIGNIFICANCE: Two secreted factors, WISP1 and WISP2, antagonistically regulate collagen linearization, and therapeutically increasing the WISP2:WISP1 ratio in tumors limits collagen linearization and inhibits metastasis.See related commentary by Barcus and Longmore, p. 5611.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/prevención & control , Proteínas CCN de Señalización Intercelular/antagonistas & inhibidores , Proteínas CCN de Señalización Intercelular/metabolismo , Colágeno Tipo I/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/prevención & control , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas CCN de Señalización Intercelular/genética , Movimiento Celular , Proliferación Celular , Colágeno Tipo I/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Invasividad Neoplásica , Pronóstico , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Transducción de Señal , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Arterioscler Thromb Vasc Biol ; 40(5): 1256-1274, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32160773

RESUMEN

OBJECTIVE: In view of our previous observations on differential expression of LMCD1 (LIM and cysteine-rich domains 1) in human versus rodents, we asked the question whether LMCD1 plays a species-specific role in the development of vascular lesions. Approach and Results: A combination of genetic, molecular, cellular, and disease models were used to test species-specific role of LMCD1 in the pathogenesis of vascular lesions. Here, we report species-specific regulation of LMCD1 expression in mediating vascular smooth muscle cell proliferation and migration during vascular wall remodeling in humans versus mice. Thrombin induced LMCD1 expression in human aortic smooth muscle cells but not mouse aortic smooth muscle cells via activation of Par1 (protease-activated receptor 1)-Gαq/11 (Gα protein q/11)-PLCß3 (phospholipase Cß3)-NFATc1 (nuclear factor of activated T cells 1) signaling. Furthermore, although LMCD1 mediates thrombin-induced proliferation and migration of both human aortic smooth muscle cells and mouse aortic smooth muscle cells via influencing E2F1 (E2F transcription factor 1)-mediated CDC6 (cell division cycle 6) expression and NFATc1-mediated IL (interleukin)-33 expression, respectively, in humans, it acts as an activator, and in mice, it acts as a repressor of these transcriptional factors. Interestingly, LMCD1 repressor activity was nullified by N-myristoyltransferase 2-mediated myristoylation in mouse. Besides, we found increased expression of LMCD1 in human stenotic arteries as compared to nonstenotic arteries. On the other hand, LMCD1 expression was decreased in neointimal lesions of mouse injured arteries as compared to noninjured arteries. CONCLUSIONS: Together, these observations reveal that LMCD1 acts as an activator and repressor of E2F1 and NFATc1 in humans and mice, respectively, in the induction of CDC6 and IL-33 expression during development of vascular lesions. Based on these findings, LMCD could be a potential target for drug development against restenosis and atherosclerosis in humans.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Factor de Transcripción E2F1/metabolismo , Interleucina-33/metabolismo , Proteínas con Dominio LIM/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Factores de Transcripción NFATC/metabolismo , Remodelación Vascular , Lesiones del Sistema Vascular/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Proteínas Co-Represoras/genética , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/genética , Femenino , Regulación de la Expresión Génica , Humanos , Interleucina-33/genética , Proteínas con Dominio LIM/genética , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Ácido Mirístico/metabolismo , Factores de Transcripción NFATC/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Especificidad de la Especie , Trombina/farmacología , Remodelación Vascular/efectos de los fármacos , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/patología
3.
Biosci Rep ; 39(12)2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31763672

RESUMEN

Oviduct-specific glycoprotein (OVGP1) is a high molecular weight chitinase-like protein belonging to GH18 family. It is secreted by non-ciliated epithelial cells of oviduct during estrous cycle providing an essential milieu for fertilization and embryo development. The present study reports the characterization of buffalo OVGP1 through structural modeling, carbohydrate-binding properties and evolutionary analysis. Structural model displayed the typical fold of GH18 family members till the boundary of chitinase-like domain further consisting of a large (ß/α)8 TIM barrel sub-domain and a small (α+ß) sub-domain. Two critical catalytic residues were found substituted in the catalytic centre (Asp to Phe118, Glu to Leu120) compared with the active chitinase. The carbohydrate-binding groove in TIM barrel was lined with various conserved aromatic residues. Molecular docking with different sugars revealed the involvement of various residues in hydrogen-bonding and non-bonded contacts. Most of the substrate-binding residues were conserved except for a few replacements (Ser13, Lys48, Asp49, Pro50, Asp167, Glu199, Gln272 and Phe275) in comparison with other GH18 members. The residues Trp10, Trp79, Asn80, Gln272, Phe275 and Trp334 were involved in recognition of all six ligands. The α+ß sub-domain participated in sugar-binding through Thr270, Gln272, Tyr242 and Phe275. The binding assays revealed significant sugar-binding with purified native and recombinant OVGP1. Phylogenetic analysis revealed that OVGP1 was closely related to AMCases followed by other CLPs and evolution of OVGP1 occurred through several gene duplications. This is the first study describing the structural characteristics of OVGP1 that will further help to understand its interaction with gametes to perform crucial reproductive functions.


Asunto(s)
Búfalos/genética , Glicoproteínas/ultraestructura , Conformación Proteica , Relación Estructura-Actividad , Animales , Dominio Catalítico/genética , Femenino , Glicoproteínas/química , Glicoproteínas/genética , Humanos , Simulación del Acoplamiento Molecular
4.
EMBO J ; 38(16): e101302, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31294477

RESUMEN

Collagen linearization is a hallmark of aggressive tumors and a key pathogenic event that promotes cancer cell invasion and metastasis. Cell-generated mechanical tension has been proposed to contribute to collagen linearization in tumors, but it is unknown whether other mechanisms play prominent roles in this process. Here, we show that the secretome of cancer cells is by itself able to induce collagen linearization independently of cell-generated mechanical forces. Among the tumor cell-secreted factors, we find a key role in this process for the matricellular protein WISP1 (CCN4). Specifically, WISP1 directly binds to type I collagen to promote its linearization in vitro (in the absence of cells) and in vivo in tumors. Consequently, WISP1-induced type I collagen linearization facilitates tumor cell invasion and promotes spontaneous breast cancer metastasis, without significantly affecting gene expression. Furthermore, higher WISP1 expression in tumors from cancer patients correlates with faster progression to metastatic disease and poor prognosis. Altogether, these findings reveal a conceptually novel mechanism whereby pro-metastatic collagen linearization critically depends on a cancer cell-secreted factor.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas CCN de Señalización Intercelular/genética , Proteínas CCN de Señalización Intercelular/metabolismo , Colágeno Tipo I/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Pronóstico , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba
5.
Arterioscler Thromb Vasc Biol ; 39(6): 1212-1226, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31043075

RESUMEN

Objective- IL (interleukin)-33 has been shown to play a role in endothelial dysfunction, but its role in atherosclerosis is controversial. Therefore, the purpose of this study is to examine its role in vascular wall remodeling following injury. Approach and Results- Thrombin induced IL-33 expression in a time-dependent manner in human aortic smooth muscle cells and inhibition of its activity by its neutralizing antibody suppressed thrombin induced human aortic smooth muscle cell migration but not DNA synthesis. In exploring the mechanisms, we found that Par1 (protease-activated receptor 1), Gαq/11 (Gα protein q/11), PLCß3 (phospholipase Cß3), NFATc1 (nuclear factor of activated T cells), E2F1 (E2F transcription factor 1), and LMCD1 (LIM and cysteine-rich domains protein 1) are involved in thrombin-induced IL-33 expression and migration. Furthermore, we identified an NFAT-binding site at -100 nt that mediates thrombin-induced IL-33 promoter activity. Interestingly, we observed that NFATc1, E2F1, and LMCD1 bind to NFAT site in response to thrombin and found that LMCD1, while alone has no significant effect, enhanced either NFATc1 or E2F1-dependent IL-33 promoter activity. In addition, we found that guidewire injury induces IL-33 expression in SMC and its neutralizing antibodies substantially reduce SMC migration and neointimal growth in vivo. Increased expression of IL-33 was also observed in human atherosclerotic lesions as compared to arteries without any lesions. Conclusions- The above findings reveal for the first time that thrombin-induced human aortic smooth muscle cell migration and injury-induced neointimal growth require IL-33 expression. In addition, thrombin-induced IL-33 expression requires LMCD1 enhanced combinatorial activation of NFATc1 and E2F1.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Factor de Transcripción E2F1/metabolismo , Interleucina-33/metabolismo , Proteínas con Dominio LIM/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Factores de Transcripción NFATC/metabolismo , Neointima , Lesiones del Sistema Vascular/metabolismo , Animales , Sitios de Unión , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Co-Represoras/genética , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/genética , Femenino , Arteria Femoral/efectos de los fármacos , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Arteria Femoral/patología , Células HEK293 , Humanos , Interleucina-33/genética , Proteínas con Dominio LIM/genética , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Factores de Transcripción NFATC/genética , Regiones Promotoras Genéticas , Transducción de Señal , Regulación hacia Arriba , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/patología
6.
F1000Res ; 7: 274, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29983921

RESUMEN

Evidence is increasing on the crucial role of the extracellular matrix (ECM) in breast cancer progression, invasion and metastasis with almost all mortality cases owing to metastasis. The epithelial-mesenchymal transition is the first signal of metastasis involving different transcription factors such as Snail, TWIST, and ZEB1. ECM remodeling is a major event promoting cancer invasion and metastasis; where matrix metalloproteinases (MMPs) such as MMP-2, -9, -11, and -14 play vital roles degrading the matrix proteins for cancer spread. The ß-D mannuronic acid (MMP inhibitor) has anti-metastatic properties through inhibition of MMP-2, and -9 and could be a potential therapeutic agent. Besides the MMPs, the enzymes such as LOXL2, LOXL4, procollagen lysyl hydroxylase-2, and heparanase also regulate breast cancer progression. The important ECM proteins like integrins (b1-, b5-, and b6- integrins), ECM1 protein, and Hic-5 protein are also actively involved in breast cancer development. The stromal cells such as tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and adipocytes also contribute in tumor development through different processes. The TAMs become proangiogenic through secretion of VEGF-A and building vessel network for nourishment and invasion of the tumor mass. The latest developments of ECM involvement in breast cancer progression has been discussed in this review and this study will help researchers in designing future work on breast cancer pathogenesis and developing therapy targeted to the ECM components.

7.
J Biol Chem ; 293(9): 3088-3103, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29326163

RESUMEN

Restenosis arises after vascular injury and is characterized by arterial wall thickening and decreased arterial lumen space. Vascular injury induces the production of thrombin, which in addition to its role in blood clotting acts as a mitogenic and chemotactic factor. In exploring the molecular mechanisms underlying restenosis, here we identified LMCD1 (LIM and cysteine-rich domains 1) as a gene highly responsive to thrombin in human aortic smooth muscle cells (HASMCs). Of note, LMCD1 depletion inhibited proliferation of human but not murine vascular smooth muscle cells. We also found that by physically interacting with E2F transcription factor 1, LMCD1 mediates thrombin-induced expression of the CDC6 (cell division cycle 6) gene in the stimulation of HASMC proliferation. Thrombin-induced LMCD1 and CDC6 expression exhibited a requirement for protease-activated receptor 1-mediated Gαq/11-dependent activation of phospholipase C ß3. Moreover, the expression of LMCD1 was highly induced in smooth muscle cells located at human atherosclerotic lesions and correlated with CDC6 expression and that of the proliferation marker Ki67. Furthermore, the LMCD1- and SMCαactin-positive cells had higher cholesterol levels in the atherosclerotic lesions. In conclusion, these findings indicate that by acting as a co-activator with E2F transcription factor 1 in CDC6 expression, LMCD1 stimulates HASMC proliferation and thereby promotes human atherogenesis, suggesting an involvement of LMCD1 in restenosis.


Asunto(s)
Aterosclerosis/metabolismo , Proteínas Co-Represoras/metabolismo , Proteínas con Dominio LIM/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Trombina/farmacología , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Ratas , Adulto Joven
8.
J Biol Chem ; 292(34): 14080-14091, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28655771

RESUMEN

Although the involvement of Rho proteins in the pathogenesis of vascular diseases is well studied, little is known about the role of their upstream regulators, the Rho guanine nucleotide exchange factors (RhoGEFs). Here, we sought to identify the RhoGEFs involved in monocyte chemotactic protein 1 (MCP1)-induced vascular wall remodeling. We found that, among the RhoGEFs tested, MCP1 induced tyrosine phosphorylation of p115 RhoGEF but not of PDZ RhoGEF or leukemia-associated RhoGEF in human aortic smooth muscle cells (HASMCs). Moreover, p115 RhoGEF inhibition suppressed MCP1-induced HASMC migration and proliferation. Consistent with these observations, balloon injury (BI) induced p115 RhoGEF tyrosine phosphorylation in rat common carotid arteries, and siRNA-mediated down-regulation of its levels substantially attenuated BI-induced smooth muscle cell migration and proliferation, resulting in reduced neointima formation. Furthermore, depletion of p115 RhoGEF levels also abrogated MCP1- or BI-induced Rac1-NFATc1-cyclin D1-CDK6-PKN1-CDK4-PAK1 signaling, which, as we reported previously, is involved in vascular wall remodeling. Our findings also show that protein kinase N1 (PKN1) downstream of Rac1-cyclin D1/CDK6 and upstream of CDK4-PAK1 in the p115 RhoGEF-Rac1-NFATc1-cyclin D1-CDK6-PKN1-CDK4-PAK1 signaling axis is involved in the modulation of vascular wall remodeling. Of note, we also observed that CCR2-Gi/o-Fyn signaling mediates MCP1-induced p115 RhoGEF and Rac1 GTPase activation. These findings suggest that p115 RhoGEF is critical for MCP1-induced HASMC migration and proliferation in vitro and for injury-induced neointima formation in vivo by modulating Rac1-NFATc1-cyclin D1-CDK6-PKN1-CDK4-PAK1 signaling.


Asunto(s)
Quimiocina CCL2/agonistas , Modelos Biológicos , Músculo Liso Vascular/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Proteína de Unión al GTP rac1/agonistas , Animales , Aorta , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común , Movimiento Celular , Proliferación Celular , Células Cultivadas , Quimiocina CCL2/metabolismo , Activación Enzimática , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Neointima/metabolismo , Neointima/patología , Fosforilación , Interferencia de ARN , Ratas , Factores de Intercambio de Guanina Nucleótido Rho/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Especificidad por Sustrato , Remodelación Vascular , Proteína de Unión al GTP rac1/metabolismo
9.
Sci Rep ; 6: 28687, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27363897

RESUMEN

Monocyte chemotactic protein 1 (MCP1) stimulates phosphorylation of cortactin on Y421 and Y446 residues in a time-dependent manner and phosphorylation at Y446 but not Y421 residue is required for MCP1-induced CDK-interacting protein 1 (p21Cip1) nuclear export and degradation in facilitating human aortic smooth muscle cell (HASMC) proliferation. In addition, MCP1-induced cortactin tyrosine phosphorylation, p21Cip1 degradation and HASMC proliferation are dependent on Fyn activation. Upstream to Fyn, MCP1 stimulated C-C chemokine receptor type 2 (CCR2) and Gi/o and inhibition of either one of these molecules using their specific antagonists or inhibitors attenuated MCP1-induced cortactin tyrosine phosphorylation, p21Cip1 degradation and HASMC proliferation. Cortactin phosphorylation at Y446 residue is also required for another G protein-coupled receptor (GPCR) agonist, thrombin-induced p21Cip1 nuclear export and its degradation in promoting HASMC proliferation. Quite interestingly, the receptor tyrosine kinase (RTK) agonist, platelet-derived growth factor-BB (PDGF-BB)-induced p21Cip1 degradation and HASMC proliferation do not require cortactin tyrosine phosphorylation. Together, these findings demonstrate that tyrosine phosphorylation of cortactin at Y446 residue is selective for only GPCR but not RTK agonist-induced nuclear export and proteolytic degradation of p21Cip1 in HASMC proliferation.


Asunto(s)
Núcleo Celular/metabolismo , Cortactina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Miocitos del Músculo Liso/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/farmacología , Cortactina/genética , Humanos , Microscopía Fluorescente , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteolisis/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Tirosina/genética , Tirosina/metabolismo
10.
J Cell Sci ; 129(6): 1234-49, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26857814

RESUMEN

Pak1 plays an important role in several cellular processes, including cell migration, but its role in pathological angiogenesis is not known. Here, we have determined its role in pathological retinal angiogenesis using an oxygen-induced retinopathy (OIR) model. VEGFA induced phosphorylation of Pak1 and its effector cofilin in a manner that was dependent on time as well as p38MAPKß (also known as MAPK11) in human retinal microvascular endothelial cells (HRMVECs). Depletion of the levels of any of these molecules inhibited VEGFA-induced HRMVEC F-actin stress fiber formation, migration, proliferation, sprouting and tube formation. In accordance with these observations, hypoxia induced Pak1 and cofilin phosphorylation with p38MAPKß being downstream to Pak1 and upstream to cofilin in mouse retina. Furthermore, Pak1 deficiency abolished hypoxia-induced p38MAPKß and cofilin phosphorylation and abrogated retinal endothelial cell proliferation, tip cell formation and neovascularization. In addition, small interfering RNA (siRNA)-mediated downregulation of p38MAPKß or cofilin levels in the wild-type mouse retina also diminished endothelial cell proliferation, tip cell formation and neovascularization. Taken together, these observations suggest that, although the p38MAPKß-Pak1-cofilin axis is required for HRMVEC migration, proliferation, sprouting and tubulogenesis, Pak1-p38MAPKß-cofilin signaling is also essential for hypoxia-induced mouse retinal endothelial cell proliferation, tip cell formation and neovascularization.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Cofilina 1/metabolismo , Neovascularización Retiniana/metabolismo , Factores Despolimerizantes de la Actina/genética , Animales , Proliferación Celular , Células Cultivadas , Cofilina 1/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/metabolismo , Retina/citología , Retina/metabolismo , Neovascularización Retiniana/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Apoptosis ; 21(2): 209-24, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26659075

RESUMEN

MGP-40 is a chitinase-like protein which is over expressed during mammary gland involution. However, its physiological function in the mammary gland is poorly understood. In the present investigation, we have reported the functional significance of buffalo specific MGP-40 in the mammary gland by using an in vitro model of the buffalo mammary epithelial cell (BuMEC) line. MGP-40 was highly up regulated in BuMECs in serum starved condition as well as after treatment with prolactin suggesting its role in the stress response. Subsequently, to study the effect of MGP-40 on BuMECs, the cells were transfected with a mammalian expression construct of pCI neo harboring MGP-40 gene. It was observed that over expression of MGP-40 enhanced proliferation of BuMECs and protected the cells from apoptosis under serum free condition. In contrast, MGP-40 attenuated the mitogenic effect of insulin in BuMECs. Besides, over expression of the MGP-40 reduced dome formation, acinar polarization and casein synthesis in BuMECs in the presence of lactogenic hormones, it also induced Stat3 phosphorylation and epithelial to mesenchymal transition (EMT) -like features. Together, our data suggest that MGP-40 is involved in protection of BuMECs under stress conditions, inhibits cellular differentiation and induces EMT-like features. A schematic diagram depicting possible association of MGP-40 in various molecular pathways has been presented.


Asunto(s)
Apoptosis , Células Epiteliales/fisiología , Glicoproteínas/metabolismo , Animales , Búfalos , Caseínas/genética , Caseínas/metabolismo , Polaridad Celular , Proliferación Celular , Forma de la Célula , Células Cultivadas , Quitinasas/genética , Quitinasas/metabolismo , Femenino , Glicoproteínas/genética , Glándulas Mamarias Animales/citología , Prolactina/fisiología , Activación Transcripcional
12.
Mol Biol Cell ; 26(25): 4589-606, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26490115

RESUMEN

Monocyte chemotactic protein 1 (MCP1) stimulates vascular smooth muscle cell (VSMC) migration in vascular wall remodeling. However, the mechanisms underlying MCP1-induced VSMC migration have not been understood. Here we identify the signaling pathway associated with MCP1-induced human aortic smooth muscle cell (HASMC) migration. MCP1, a G protein-coupled receptor agonist, activates phosphorylation of cortactin on S405 and S418 residues in a time-dependent manner, and inhibition of its phosphorylation attenuates MCP1-induced HASMC G-actin polymerization, F-actin stress fiber formation, and migration. Cortactin phosphorylation on S405/S418 is found to be critical for its interaction with WAVE2, a member of the WASP family of cytoskeletal regulatory proteins required for cell migration. In addition, the MCP1-induced cortactin phosphorylation is dependent on PLCß3-mediated PKCδ activation, and siRNA-mediated down-regulation of either of these molecules prevents cortactin interaction with WAVE2, affecting G-actin polymerization, F-actin stress fiber formation, and HASMC migration. Upstream, MCP1 activates CCR2 and Gαq/11 in a time-dependent manner, and down-regulation of their levels attenuates MCP1-induced PLCß3 and PKCδ activation, cortactin phosphorylation, cortactin-WAVE2 interaction, G-actin polymerization, F-actin stress fiber formation, and HASMC migration. Together these findings demonstrate that phosphorylation of cortactin on S405 and S418 residues is required for its interaction with WAVE2 in MCP1-induced cytoskeleton remodeling, facilitating HASMC migration.


Asunto(s)
Movimiento Celular/genética , Quimiocina CCL2/metabolismo , Cortactina/metabolismo , Músculo Liso Vascular/crecimiento & desarrollo , Fosfolipasa C beta/metabolismo , Actinas/genética , Actinas/metabolismo , Aorta/crecimiento & desarrollo , Aorta/metabolismo , Quimiocina CCL2/genética , Cortactina/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Humanos , Fibras Musculares Esqueléticas/metabolismo , Músculo Liso Vascular/metabolismo , Fosfolipasa C beta/genética , Fosforilación , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo
13.
J Proteomics ; 119: 100-11, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25661041

RESUMEN

Mammary gland is an exocrine and sebaceous gland made up of branching network of ducts that end in alveoli. Milk is synthesized in the alveoli and secreted into alveolar lumen. Mammary gland represents an ideal system for the study of organogenesis that undergoes successive cycles of pregnancy, lactation and involution. To gain insights on the molecular events that take place in pubertal and lactating mammary gland, we have identified 43 differentially expressed proteins in mammary tissue of heifer (non-lactating representing a virgin mammary gland), and lactating buffaloes (Bubalus bubalis) by 2D-difference gel electrophoresis (2D-DIGE) and mass spectrometry. Twenty one proteins were upregulated during lactation whereas 8 proteins were upregulated in heifer mammary gland significantly (p<0.05). Bioinformatics analyses of the identified proteins showed that a majority of the proteins are involved in metabolic processes. The differentially expressed proteins were validated by real-time PCR and Western blotting. We observed differential expressions of certain new proteins including EEF1D, HSPA5, HSPD1 and PRDX6 during lactation which have not been reported before. The differentially expressed proteins were mapped to available biological pathways and networks involved in lactation. This study signifies the importance of some proteins which are preferentially expressed during lactation and in heifer mammary gland. BIOLOGICAL SIGNIFICANCE: This work is important because we have generated information in water buffalo (B. bubalis) for the first time which is the major milk producing animal in Indian Subcontinent. Out of a present production of 133milliontons of milk produced in India, contribution of buffalo milk is around 54%. Its physiology is somewhat different from the lactating cows. Buffalo milk composition varies from cow milk in terms of higher fat and total solid content, which confers an advantage in preparation of specialized cheese, curd and other dairy products. Being a major milk producing animal in India it is highly essential to understand the lactation associated proteins in the mammary gland of buffalo. In the present investigation our attempt has been to identify new protein evidences which are expressed in lactating buffalo mammary gland and have not been reported before. The findings reported in the present study will help in understanding the lactation biology of buffalo mammary gland in particular and the mammary gland biology in general.


Asunto(s)
Búfalos/metabolismo , Regulación de la Expresión Génica/fisiología , Lactancia/fisiología , Glándulas Mamarias Animales/metabolismo , Embarazo/metabolismo , Proteoma/metabolismo , Animales , Femenino
14.
Int J Nanomedicine ; 9: 5177-87, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25429214

RESUMEN

Infection-related complications have been a critical issue for the application of titanium orthopedic implants. The use of Ag nanoparticles offers a potential approach to incorporate antimicrobial properties into the titanium implants. In this work, a novel and simple method was developed for synthesis of Ag (II) oxide deposited TiO2 nanotubes (TiNTs) using electrochemical anodization followed by Ag electroplating processes in the same electrolyte. The quantities of AgO nanoparticles deposited in TiNT were controlled by selecting different electroplating times and voltages. It was shown that AgO nanoparticles were crystalline and distributed throughout the length of the nanotubes. Inductively coupled plasma mass spectrometry tests showed that the quantities of released Ag were less than 7 mg/L after 30 days at 37°C. Antimicrobial assay results show that the AgO-deposited TiNTs can effectively kill the Escherichia coli bacteria. Although the AgO-deposited TiNTs showed some cytotoxicity, it should be controllable by optimization of the electroplating parameters and incorporation of cell growth factor. The results of this study indicated that antimicrobial properties could be added to nanotextured medical implants through a simple and cost effective method.


Asunto(s)
Antiinfecciosos/química , Nanopartículas del Metal/química , Nanotubos/química , Óxidos/química , Compuestos de Plata/química , Titanio/química , Animales , Antiinfecciosos/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Técnicas Electroquímicas , Escherichia coli/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Nanotecnología , Óxidos/farmacología , Compuestos de Plata/farmacología , Titanio/farmacología
15.
PLoS One ; 9(8): e102515, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25111801

RESUMEN

Mammary gland is made up of a branching network of ducts that end with alveoli which surrounds the lumen. These alveolar mammary epithelial cells (MEC) reflect the milk producing ability of farm animals. In this study, we have used 2D-DIGE and mass spectrometry to identify the protein changes in MEC during immediate early, peak and late stages of lactation and also compared differentially expressed proteins in MEC isolated from milk of high and low milk producing cows. We have identified 41 differentially expressed proteins during lactation stages and 22 proteins in high and low milk yielding cows. Bioinformatics analysis showed that a majority of the differentially expressed proteins are associated in metabolic process, catalytic and binding activity. The differentially expressed proteins were mapped to the available biological pathways and networks involved in lactation. The proteins up-regulated during late stage of lactation are associated with NF-κB stress induced signaling pathways and whereas Akt, PI3K and p38/MAPK signaling pathways are associated with high milk production mediated through insulin hormone signaling.


Asunto(s)
Lactancia , Glándulas Mamarias Animales/metabolismo , Leche/metabolismo , Proteómica , Electroforesis Bidimensional Diferencial en Gel , Animales , Bovinos , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Proteínas de la Leche/genética , Proteínas de la Leche/metabolismo , Análisis de Componente Principal
16.
Macromol Biosci ; 14(6): 853-71, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24610743

RESUMEN

A robust self-assembly of nanoparticles into fibers and 3D scaffolds is designed and fabricated by functionalizing a RAFT-polymerized amphiphilic triblock copolymer with designer ionic complementary peptides so that the assembled core-shell polymeric nanoparticles are directed by peptide assembly into continuous "nanoparticle fibers," ultimately leading to 3D fiber scaffolds. The assembled nanostructure is confirmed by FESEM and optical microscopy. The assembly is not hindered when a protein (insulin) is incorporated within the nanoparticles as an active ingredient. MTS cytotoxicity tests on SW-620 cell lines show that the peptides, copolymers, and peptide-copolymer conjugates are biocompatible. The methodology of self-assembled nanoparticle fibers and 3D scaffolds is intended to combine the advantages of a flexible hydrogel scaffold with the versatility of controlled release nanoparticles to offer unprecedented ability to incorporate desired drug(s) within a self-assembled scaffold system with individual control over the release of each drug.


Asunto(s)
Hipoglucemiantes , Insulina , Nanofibras/química , Péptidos/química , Línea Celular , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/farmacología , Insulina/química , Insulina/farmacología , Ensayo de Materiales
17.
J Mater Chem B ; 2(28): 4500-4508, 2014 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-32261552

RESUMEN

Four near-infrared fluorescent probes (A, B, C and D) have been synthesized, characterized, and evaluated for detection of lysosomal pH inside living cells. The fluorescent probes display highly sensitive and selective fluorescent response to acidic pH as the acidic pH results in drastic structural changes from spirocyclic (non-fluorescent) forms to ring-opening (fluorescent) forms of the fluorescent probes. The fluorescence intensities of the fluorescent probes (B, C and D) increase significantly by more than 200-fold from pH 7.4 to 4.2. The fluorescent probe D bearing the N-(2-hydroxyethyl) ethylene amide residue possesses the advantages of high sensitivity, excellent photostability, good cell membrane permeability, strong pH dependence, and low auto-fluorescence background. It has been successfully applied to selectively stain lysosomes and detect lysosomal pH changes inside normal endothelial and breast cancer cells.

18.
Genomics Proteomics Bioinformatics ; 11(6): 335-44, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24296084

RESUMEN

Preeclampsia, a complication of pregnancy characterized by hypertension and proteinuria, has been found to reduce the subsequent risk for breast cancer in female offspring. As this protective effect could be due to exposure to preeclampsia-specific proteins during intrauterine life, the proteomic profiles of umbilical cord blood plasma between preeclamptic and normotensive pregnancies were compared. Umbilical cord plasma samples, depleted of 14 abundant proteins, were subjected to proteomic analysis using the quantitative method of nanoACQUITY ultra performance liquid chromatography-mass spectrometry with elevated energy mode of acquisition(E) (NanoUPLC-MS(E)). Sixty-nine differentially expressed proteins were identified, of which 15 and 6 proteins were only detected in preeclamptic and normotensive pregnancies, respectively. Additionally, expression of 8 proteins (gelsolin, complement C5, keratin type I cytoskeletal 10, pigment epithelium-derived factor, complement factor B, complement component C7, hemoglobin subunit gamma-2 and alpha-fetoprotein) were up-regulated in preeclampsia with a fold change of ≥2.0 when compared to normotensive pregnancies. The identification of alpha-fetoprotein in preeclamptic umbilical cord blood plasma supported the validity of this screen as alpha-fetoprotein has anti-estrogenic properties and has previously been linked to preeclampsia as well as a reduced breast cancer risk. The findings of this pilot study may provide new insights into the mechanistic link between preeclampsia and potentially reduced breast cancer susceptibility in adult life.


Asunto(s)
Proteínas Sanguíneas/análisis , Neoplasias de la Mama/sangre , Sangre Fetal/química , Preeclampsia/sangre , Proteómica , Neoplasias de la Mama/química , Cromatografía Liquida , Susceptibilidad a Enfermedades , Femenino , Sangre Fetal/metabolismo , Humanos , Espectrometría de Masas , Proyectos Piloto , Embarazo
19.
Proteomics ; 13(21): 3189-204, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24030930

RESUMEN

Mammary gland is made up of a branching network of ducts that end in alveoli. Terminally differentiated mammary epithelial cells (MECs) constitute the innermost layer of aveoli. They are milk-secreting cuboidal cells that secrete milk proteins during lactation. Little is known about the expression profile of proteins in the metabolically active MECs during lactation or their functional role in the lactation process. In the present investigation, we have reported the proteome map of MECs in lactating cows using 2DE MALDI-TOF/TOF MS and 1D-Gel-LC-MS/MS. MECs were isolated from milk using immunomagnetic beads and confirmed by RT-PCR and Western blotting. The 1D-Gel-LC-MS/MS and 2DE-MS/MS based approaches led to identification of 431 and 134 proteins, respectively, with a total of 497 unique proteins. Proteins identified in this study were clustered into functional groups using bioinformatics tools. Pathway analysis of the identified proteins revealed 28 pathways (p < 0.05) providing evidence for involvement of various proteins in lactation function. This study further provides experimental evidence for the presence of many proteins that have been predicted in annotated bovine genome. The data generated further provide a set of bovine MEC-specific proteins that will help the researchers to understand the molecular events taking place during lactation.


Asunto(s)
Células Epiteliales/química , Glándulas Mamarias Animales/citología , Leche/citología , Proteoma/análisis , Animales , Bovinos , Femenino , Lactancia/metabolismo , Glándulas Mamarias Animales/metabolismo , Redes y Vías Metabólicas , Mapas de Interacción de Proteínas , Proteínas/análisis , Proteínas/química , Proteínas/metabolismo , Proteoma/química
20.
Anal Chim Acta ; 758: 138-44, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23245906

RESUMEN

Near-infrared emissive BODIPY polymeric dye bearing cancer-homing cyclic arginine-glycine-aspartic acid (RGD) peptide residues (polymer B) was prepared by post-polymerization functionalization of BODIPY polymeric dye bearing bromo groups through tetra(ethylene glycol) tethered spacers (polymer A) with thiol-functionalized RGD cancer-homing peptide through thioether bonds under a mild basic condition. Polymer B possesses excellent water solubility, good photostability, biocompatibility and resistance to nonspecific interactions to normal endothelial cells, and can efficiently detect breast tumor cells through specific cooperative binding of cancer-homing RGD peptides to αvß3 integrins of cancer cells while its parent polymer A without RGD residues fails to target cancer cells.


Asunto(s)
Compuestos de Boro , Neoplasias de la Mama/diagnóstico , Colorantes Fluorescentes , Oligopéptidos , Polímeros , Compuestos de Boro/química , Compuestos de Boro/metabolismo , Mama/citología , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Oligopéptidos/química , Oligopéptidos/metabolismo , Imagen Óptica , Polímeros/química , Polímeros/metabolismo , Solubilidad , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA