Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(1)2022 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-36613857

RESUMEN

Vasculogenic properties of bone marrow-derived mesenchymal stem cells (MSCs) have been reported, but it is still unclear whether the vasculogenic properties are restricted to some populations of MSCs or whether the entire population of MSCs has these properties. We cultured two different populations of MSCs in different culture media and their vasculogenic properties were evaluated using In vitro spheroid sprouting assay. Neither population of MSCs expressed markers of endothelial progenitor cells (EPCs), but they were different in the profiling of angiogenic factor expression as well as vasculogenic properties. One population of MSCs expressed basic fibroblast growth factor (bFGF) and another expressed hepatocyte growth factor (HGF). MSCs expressing HGF exhibited In vitro angiogenic sprouting capacity in response to bFGF derived from other MSCs as well as to their autocrine HGF. The vasculogenic mesenchymal stem cells (vMSCs) derived from the bone marrow also enhanced In vitro angiogenic sprouting capacity of human umbilical vein endothelial cells (HUVECs) in an HGF-dependent manner. These results suggest that MSCs exhibit different vasculogenic properties, and vMSCs that are different from EPCs may contribute to neovascularization and could be a promising cellular therapy for cardiovascular diseases.


Asunto(s)
Médula Ósea , Células Madre Mesenquimatosas , Humanos , Células Endoteliales de la Vena Umbilical Humana , Neovascularización Fisiológica/fisiología , Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas
2.
Am J Physiol Heart Circ Physiol ; 320(4): H1403-H1410, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33577432

RESUMEN

Excessive vascular permeability occurs in inflammatory disease processes. Vascular endothelial cadherin (VE-cadherin) is an adhesion protein that controls vascular permeability. We identified monoclonal antibodies (mAbs) to human VE-cadherin that activate cell adhesion and inhibit the increased permeability of endothelial cell monolayers induced by thrombin receptor activator peptide-6 (TRAP-6). Two mAbs, 8A12c and 3A5a, reduce permeability, whereas an inhibitory mAb, 2E11d, enhances permeability. Activating mAbs also reduce permeability induced by tumor necrosis factor-α (TNF-α) and vascular endothelial cell growth factor (VEGF). The activating mAbs also stabilize the organization of the adherens junctions that are disrupted by TRAP-6, VEGF, or TNF-α. The activating mAbs act directly on the adhesive function of VE-cadherin because they did not block the accumulation of actin filaments stimulated by TRAP-6 and enhance physical cell-cell adhesion of VE-cadherin-expressing tissue culture cells. Therefore, VE-cadherin function can be regulated at the cell surface to control endothelial permeability.NEW & NOTEWORTHY Excessive vascular permeability is a serious complication of many inflammatory disease conditions. We have developed monoclonal antibodies that inhibit increases in endothelial monolayer permeability induced by several signaling factors by activating VE-cadherin mediated adhesion and stabilizing cell junctions. These antibodies and/or the mechanisms they reveal may lead to important therapeutics to treat vascular leakiness and inflammation.


Asunto(s)
Uniones Adherentes/efectos de los fármacos , Anticuerpos Monoclonales/farmacología , Cadherinas/agonistas , Permeabilidad Capilar/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Uniones Adherentes/metabolismo , Antígenos CD/metabolismo , Cadherinas/antagonistas & inhibidores , Cadherinas/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Nocodazol/farmacología , Oligopéptidos/farmacología , Receptores de Trombina/agonistas , Receptores de Trombina/metabolismo , Transducción de Señal , Moduladores de Tubulina/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
3.
Biochem Biophys Res Commun ; 522(4): 917-923, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-31806373

RESUMEN

Diabetes induces cellular dysfunction in dermal fibroblasts, such as impairment in migration, which is a major cause of chronic wound. Here, we demonstrated that the migration of human dermal fibroblasts was impaired under a high glucose culture condition. Substance P (SP) rescued the impaired migration of the fibroblasts. The activity of Rac1, Rho-associated kinase (ROCK), and Src was required for SP-mediated rescue of fibroblast migration. SP activated Rac1 and Src, whereas, NSC23766, a Rac1 inhibitor, and PP1 and PP2, Src inhibitors, inhibited SP-mediated enhancement of fibroblast migration. Y-27632, a ROCK inhibitor, inhibited the SP-mediated rescue of fibroblast migration. Senescence-associated ß-galactosidase activity increased in human dermal fibroblasts cultured in a high glucose environment, but SP inhibited the ß-galactosidase activity of the fibroblasts. These results suggest that SP promotes the migration of human dermal fibroblasts in diabetic-condition-mimicking cultures via the activity of Rac1, ROCK, and Src, and inhibits fibroblast senescence in hyperglycemic cultures.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Dermis/patología , Fibroblastos/patología , Hiperglucemia/patología , Sustancia P/farmacología , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Glucosa/toxicidad , Humanos , Proteína de Unión al GTP rac1/metabolismo , Quinasas Asociadas a rho/metabolismo , Familia-src Quinasas/metabolismo
4.
Int J Mol Sci ; 21(22)2020 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-33202862

RESUMEN

Endogenous bone marrow-derived mesenchymal stem cells are mobilized to peripheral blood and injured tissues in response to changes in the expression of various growth factors and cytokines in the injured tissues, including substance P (SP), transforming growth factor-beta (TGF-ß), and stromal cell-derived factor-1 (SDF-1). SP, TGF-ß, and SDF-1 are all known to induce the migration of bone marrow-derived mesenchymal stem cells (BM-MSCs). However, it is not yet clear how these stimuli influence or interact with each other during BM-MSC mobilization. This study used mouse bone marrow-derived mesenchymal stem cell-like ST2 cells and human BM-MSCs to evaluate whether SP, TGF-ß, and SDF-1 mutually regulate their respective effects on the mobilization of BM-MSCs. SP pretreatment of ST2 and BM-MSCs impaired their response to TGF-ß while the introduction of SP receptor antagonist restored the mobilization of ST2 and BM-MSCs in response to TGF-ß. TGF-ß pretreatment did not affect the migration of ST2 and BM-MSCs in response to SP, but downregulated their migration in response to SDF-1. SP pretreatment modulated the activation of TGF-ß noncanonical pathways in ST2 cells and BM-MSCs, but not canonical pathways. These results suggest that the migration of mesenchymal stem cells is regulated by complex functional interactions between SP, TGF-ß, and SDF-1. Thus, understanding the complex functional interactions of these chemotactic stimuli would contribute to ensuring the development of safe and effective combination treatments for the mobilization of BM-MSCs.


Asunto(s)
Células de la Médula Ósea/inmunología , Quimiotaxis/inmunología , Células Madre Mesenquimatosas/inmunología , Transducción de Señal/inmunología , Animales , Células de la Médula Ósea/citología , Línea Celular , Quimiocina CXCL12/inmunología , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Factor de Crecimiento Transformador beta/inmunología
5.
Biochem Biophys Res Commun ; 508(2): 361-367, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30502084

RESUMEN

Endogenous bone marrow-derived mesenchymal stem cells (BM-MSCs) are mobilized into peripheral blood and injured tissues by various growth factors and cytokines that are expressed in the injured tissues, such as substance P (SP), stromal cell derived factor-1 (SDF-1), and transforming growth factor-beta (TGF-ß). Extracellular bioactive lipid metabolites such as ceramide-1-phosphate and sphingosine-1-phosphate also modulate BM-MSC migration as SP, SDF-1, and TGF-ß. However, the roles of intrinsic lipid kinases of BM-MSCs in the stem cell migration are unclear. Here, we demonstrated that ceramide kinase mediates the chemotactic migration of BM-MSCs in response to SP, SDF-1, or TGF-ß. Furthermore, a specific inhibitor of ceramide kinase inhibited TGF-ß-induced migration of BM-MSCs and N-cadherin that is necessary for BM-MSCs migration in response to TGF-ß. Therefore, these results suggest that the intracellular ceramide kinase is required for the BM-MSCs migration and the roles of the intrinsic ceramide kinase in the migration are associated with N-cadherin regulation.


Asunto(s)
Movimiento Celular/fisiología , Células Madre Mesenquimatosas/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Antígenos CD/genética , Cadherinas/genética , Línea Celular , Ceramidas/metabolismo , Quimiocina CXCL12/metabolismo , Quimiotaxis/fisiología , Regulación de la Expresión Génica , Humanos , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sustancia P/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
6.
J Cell Physiol ; 233(1): 201-213, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28213973

RESUMEN

Transforming growth factor-beta (TGF-ß) induces the migration and mobilization of bone marrow-derived mesenchymal stem cells (BM-MSCs) to maintain bone homeostasis during bone remodeling and facilitate the repair of peripheral tissues. Although many studies have reported the mechanisms through which TGF-ß mediates the migration of various types of cells, including cancer cells, the intrinsic cellular mechanisms underlying cellular migration, and mobilization of BM-MSCs mediated by TGF-ß are unclear. In this study, we showed that TGF-ß activated noncanonical signaling molecules, such as Akt, extracellular signal-regulated kinase 1/2 (ERK1/2), focal adhesion kinase (FAK), and p38, via TGF-ß type I receptor in human BM-MSCs and murine BM-MSC-like ST2 cells. Inhibition of Rac1 by NSC23766 and Src by PP2 resulted in impaired TGF-ß-mediated migration. These results suggested that the Smad-independent, noncanonical signals activated by TGF-ß were necessary for migration. We also showed that N-cadherin-dependent intercellular interactions were required for TGF-ß-mediated migration using functional inhibition of N-cadherin with EDTA treatment and a neutralizing antibody (GC-4 antibody) or siRNA-mediated knockdown of N-cadherin. However, N-cadherin knockdown did not affect the global activation of noncanonical signals in response to TGF-ß. Therefore, these results suggested that the migration of BM-MSCs in response to TGF-ß was mediated through N-cadherin and noncanonical TGF-ß signals.


Asunto(s)
Antígenos CD/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Cadherinas/metabolismo , Movimiento Celular/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Animales , Antígenos CD/genética , Células de la Médula Ósea/metabolismo , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Neuropéptidos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/agonistas , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Familia-src Quinasas/metabolismo
7.
Biochem Biophys Res Commun ; 499(4): 960-966, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29626466

RESUMEN

Preservation of pancreatic ß-cells is required for the development of therapies for type 1 and type 2 diabetes (T1D and T2D, respectively). Our previous study demonstrated that substance P (SP) preserves ß-cell populations in mice with streptozotocin-induced T1D. Here, we demonstrated that chronic systemic treatment with SP restored the mass of ß-cells both in nonobese diabetic (NOD) mice with T1D or db/db mice with T2D. SP delayed the onset of T1D in NOD mice via immune modulation. SP inhibited immune infiltration into islets and the salivary glands of NOD mice. In db/db mice, SP treatment rescued glucose intolerance. Moreover, SP inhibited apoptosis, as well as the activation of pancreatic stellate cells in pancreatic islets of db/db mice. SP downregulated the number of α-smooth muscle actin (α-SMA) expressing cells in db/db pancreatic islets. Cleaved-caspase-3 expression was reduced in islets of SP-treated db/db mice compared to that in the control. Therefore, these results suggested that SP may preserve pancreatic ß-cells through immune modulation and protection from the stimulated activation of pancreatic stellate cells and apoptosis in T1D and T2D, respectively.


Asunto(s)
Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/patología , Células Secretoras de Insulina/patología , Sustancia P/farmacología , Animales , Apoptosis/efectos de los fármacos , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Femenino , Hiperglucemia/complicaciones , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/patología , Inmunomodulación/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Endogámicos NOD , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/patología , Sustancia P/uso terapéutico
8.
Exp Dermatol ; 27(4): 396-402, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28833499

RESUMEN

Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterized by intense pruritus and eczematous lesion. In this study, topically applied substance P (SP) significantly alleviated AD-like clinical symptoms in 2, 4, 6-trinitrochlorobenzene (TNCB)-induced dermatitis in NC/Nga mice. This effect was nullified by pretreatment of the neurokinin-1 receptor (NK-1R) antagonist CP99994. SP treatment significantly reduced the infiltration of mast cells and CD3-positive T cells as well as inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and thymic stromal lymphopoietin (TSLP), in AD-like skin lesions and decreased the levels of IgE and thymus and activation-regulated chemokine in serum. This SP-induced alleviation of allergic inflammatory responses was also confirmed as reduced activation in the axillary lymph nodes (aLN) and spleen, suggesting the systemic effect of SP on immune responses in TNCB-induced NC/Nga mice. Furthermore, SP-mediated TSLP reduction was confirmed in human keratinocyte culture under pro-inflammatory TNF-α stimulation. Taken together, these results suggest that topically administered SP may have potential as a medication for atopic dermatitis.


Asunto(s)
Degranulación de la Célula/efectos de los fármacos , Dermatitis Atópica/tratamiento farmacológico , Mastocitos/fisiología , Neurotransmisores/uso terapéutico , Sustancia P/uso terapéutico , Administración Cutánea , Animales , Complejo CD3/metabolismo , Células Cultivadas , Quimiocina CCL17/sangre , Citocinas/metabolismo , Dermatitis Atópica/inducido químicamente , Dermatitis Atópica/inmunología , Dermatitis Atópica/patología , Humanos , Inmunoglobulina E/sangre , Queratinocitos/metabolismo , Masculino , Mastocitos/patología , Ratones , Antagonistas del Receptor de Neuroquinina-1/farmacología , Neurotransmisores/farmacología , Cloruro de Picrilo , Sustancia P/farmacología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Linfopoyetina del Estroma Tímico
9.
Biochem Biophys Res Commun ; 491(4): 958-965, 2017 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-28754588

RESUMEN

Preservation of the pancreatic ß-cell population is required for the development of therapies for diabetes, which is caused by a decrease in ß-cells. Here, we demonstrate the antidiabetic effects of substance P (SP) in type 1 diabetes (T1D) mice induced with streptozotocin. SP enhanced the compensatory proliferation of ß-cells in order to restore ß-cells in response to acute injury induced by a single high-dose of streptozotocin. However, SP affected neither the basal proliferation of ß-cells nor their apoptosis. In vitro studies by using the INS-1 pancreatic ß-cell line showed that SP mediated the increase in the proliferation of ß-cells via the activation of Akt. Chronic systemic treatment with SP restored the mass of ß-cells and inhibited insulitis in T1D mice induced with multiple low-doses of streptozotocin. Therefore, systemic treatment with SP may be a promising therapeutic strategy for treating diabetes in patients with loss of functional ß-cells.


Asunto(s)
Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/prevención & control , Células Secretoras de Insulina/efectos de los fármacos , Pancreatitis/prevención & control , Sustancia P/farmacología , Enfermedad Aguda , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Pancreatitis/inducido químicamente , Pancreatitis/patología , Estreptozocina/administración & dosificación , Relación Estructura-Actividad
10.
Wound Repair Regen ; 24(2): 402-10, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-26749197

RESUMEN

Wound healing is essential for the survival and tissue homeostasis of unicellular and multicellular organisms. The current study demonstrated that the neuropeptide substance P (SP) accelerated the wound healing process, particularly in the skin. Subcutaneous treatment of SP accelerated wound closing, increased the population of α-smooth muscle actin positive myofibroblasts, and increased extracellular matrix deposition at the wound site. Moreover, SP treatment enhances angiogenesis without a local increase in the expression levels of vascular endothelial growth factor and stromal cell-derived factor-1. Importantly, SP treatment increased both the population of circulating endothelial progenitor cells in the peripheral blood and in CD31 positive cells in Matrigel plugs. The tube forming potential of endothelial cells was also enhanced by SP treatment. The results suggested that the subcutaneous injection of SP accelerated the wound healing in the skin via better reconstitution of blood vessels, which possibly followed an increase in the systemic mobilization of endothelial progenitor cells and a more effective assembly of endothelial cells into tubes.


Asunto(s)
Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/patología , Neurotransmisores/farmacología , Sustancia P/farmacología , Cicatrización de Heridas/efectos de los fármacos , Heridas y Lesiones/tratamiento farmacológico , Heridas y Lesiones/patología , Animales , Movimiento Celular , Quimiocina CXCL12 , Colágeno/farmacología , Modelos Animales de Enfermedad , Combinación de Medicamentos , Matriz Extracelular/patología , Inmunohistoquímica , Laminina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Proteoglicanos/farmacología , Factor A de Crecimiento Endotelial Vascular
11.
Biochem Biophys Res Commun ; 459(3): 504-8, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25747715

RESUMEN

Cadherin-6B induces bone morphogenetic protein (BMP) signaling to promote the epithelial mesenchymal transition (EMT) in the neural crest. We have previously found that knockdown of Cadherin-6B inhibits both BMP signaling and the emigration of the early pre-migratory neural crest cells from the dorsal neural tube. In this study, we found that inhibition of BMP signaling in the neural tube, mediated by the ectopic expression of Smad-6 or Noggin, decreased the size of the Islet-1-positive dorsal cell population. Knockdown or loss of function of Cadherin-6B suppressed the generation of Islet-1-expressing cells in the dorsal neural tube, but not the Lim-1/2 positive dorsal cell population. Our results thus indicate that Cadherin-6B is necessary for the generation of Islet-1-positive dorsal interneurons, as well as the initiation of pre-migratory neural crest cell emigration.


Asunto(s)
Proteínas Aviares/metabolismo , Cadherinas/metabolismo , Interneuronas/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas Aviares/antagonistas & inhibidores , Proteínas Aviares/genética , Proteínas Morfogenéticas Óseas/metabolismo , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Proteínas Portadoras/metabolismo , Movimiento Celular , Embrión de Pollo , Transición Epitelial-Mesenquimal , Técnicas de Silenciamiento del Gen , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Tubo Neural/citología , Tubo Neural/embriología , Tubo Neural/metabolismo , Transducción de Señal , Proteína smad6/metabolismo
12.
Tumour Biol ; 36(5): 3549-56, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25542234

RESUMEN

Epithelial-mesenchymal transition (EMT) of mammary epithelial cells is important in both normal morphogenesis of mammary glands and metastasis of breast cancer. Cadherin switching from E-cadherin to N-cadherin plays important roles in EMT. We found that cadherin switching is important in bone morphogenetic protein 4 (BMP4)-induced EMT in MCF-10A cells. BMP4 increased the phosphorylation of SMAD proteins in MCF-10A cells. Canonical BMP4 signaling decreased the expression of E-cadherin and disrupted the polarity of the tight junction protein ZO-1 in MCF-10A cells. However, the expression of N-cadherin and SNAI2 was up-regulated in BMP4-treated MCF-10A cells. MCF-10A cells that expressed N-cadherin migrated into type I collagen gels in response to BMP4 when evaluated using three-dimensional culture assays. Thus, active canonical BMP4 signaling is important for the migration and EMT of mammary epithelial cells. Moreover, the decrease in E-cadherin and/or increase in N-cadherin may be required for BMP4-induced migration and EMT.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Neoplasias de la Mama/genética , Cadherinas/biosíntesis , Transición Epitelial-Mesenquimal/genética , Neoplasias de la Mama/patología , Cadherinas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Glándulas Mamarias Humanas/patología , Morfogénesis/genética , Metástasis de la Neoplasia , Proteína de la Zonula Occludens-1/genética
13.
Cell Death Discov ; 10(1): 19, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38212369

RESUMEN

Mesenchymal stem cells are recruited from the bone marrow into breast tumors, contributing to the creation of a tumor microenvironment that fosters tropism for breast tumors. However, the intrinsic mechanisms underlying the recruitment of bone marrow-derived mesenchymal stem cells (MSCs) into the breast tumor microenvironment are still under investigation. Our discoveries identified zonula occludens-1 (ZO-1) as a specific intrinsic molecule that plays a vital role in mediating the collective migration of MSCs towards breast tumor cells and transforming growth factor beta (TGF-ß), which is a crucial factor secreted by breast tumor cells. Upon migration in response to MDA-MB-231 cells and TGF-ß, MSCs showed increased formation of adherens junction-like structures (AJs) expressing N-cadherin and α-catenin at their cell-cell contacts. ZO-1 was found to be recruited into the AJs at the cell-cell contacts between MSCs. Additionally, ZO-1 collaborated with α-catenin to regulate AJ formation, dependently on the SH3 and GUK domains of the ZO-1 protein. ZO-1 knockdown led to the impaired migration of MSCs in response to the stimuli and subsequent downregulation of AJs formation at the cell-cell contacts during MSCs migration. Overall, our study highlights the novel role of ZO-1 in guiding MSC migration towards breast tumor cells, suggesting its potential as a new strategy for controlling and re-engineering the breast tumor microenvironment.

14.
FEBS Open Bio ; 14(7): 1192-1204, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38719785

RESUMEN

Glioblastoma recruits various nontransformed cells from distant tissues. Although bone marrow-derived mesenchymal stem cells (MSCs) have been observed migrating to glioblastoma, the underlying mechanism driving MSC migration toward glioblastoma remains unclear. Tumor vascularity is critical in the context of recurrent glioblastoma and is closely linked to the expression of stromal cell-derived factor-1 (SDF-1). We demonstrated that cadherin-6 mediated MSC migration both toward SDF-1 and toward glioblastoma cells. Cadherin-6 knockdown resulted in the downregulation of MSCs capacity to migrate in response to SDF-1. Furthermore, MSCs with cadherin-6 knockdown exhibited impaired migration in response to conditioned media derived from glioblastoma cell lines (U87 and U373) expressing SDF-1, thus simulating the glioblastoma microenvironment. Moreover, MSCs enhanced the vasculogenic capacity of U87 cells without increasing the proliferation, cancer stem cell characteristics, or migration of U87. These results suggest that the current strategy of utilizing MSCs as carriers for antiglioblastoma drugs requires careful examination. Furthermore, cadherin-6 may represent a novel potential target for controlling the recruitment of MSCs toward glioblastoma.


Asunto(s)
Cadherinas , Movimiento Celular , Quimiocina CXCL12 , Glioblastoma , Células Madre Mesenquimatosas , Humanos , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/genética , Células Madre Mesenquimatosas/metabolismo , Cadherinas/metabolismo , Cadherinas/genética , Movimiento Celular/genética , Quimiocina CXCL12/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Microambiente Tumoral
15.
Dev Biol ; 366(2): 232-43, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22537493

RESUMEN

We previously provided evidence that cadherin-6B induces de-epithelialization of the neural crest prior to delamination and is required for the overall epithelial mesenchymal transition (EMT). Furthermore, de-epithelialization induced by cadherin-6B was found to be mediated by BMP receptor signaling independent of BMP. We now find that de-epithelialization is mediated by non-canonical BMP signaling through the BMP type II receptor (BMPRII) and not by canonical Smad dependent signaling through BMP Type I receptor. The LIM kinase/cofilin pathway mediates non-canonical BMPRII induced de-epithelialization, in response to either cadherin-6B or BMP. LIMK1 induces de-epithelialization in the neural tube and dominant negative LIMK1 decreases de-epithelialization induced by either cadherin-6B or BMP. Cofilin is the major known LIMK1 target and a S3A phosphorylation deficient mutated cofilin inhibits de-epithelialization induced by cadherin-6B as well as LIMK1. Importantly, LIMK1 as well as cadherin-6B can trigger ectopic delamination when co-expressed with the competence factor SOX9, showing that this cadherin-6B stimulated signaling pathway can mediate the full EMT in the appropriate context. These findings suggest that the de-epithelialization step of the neural crest EMT by cadherin-6B/BMPRII involves regulation of actin dynamics via LIMK/cofilin.


Asunto(s)
Proteínas Aviares/fisiología , Cadherinas/fisiología , Transición Epitelial-Mesenquimal/fisiología , Cresta Neural/fisiología , Factores Despolimerizantes de la Actina/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/fisiología , Embrión de Pollo , Ectodermo/citología , Ectodermo/fisiología , Quinasas Lim/fisiología , Cresta Neural/citología , Transducción de Señal
16.
Development ; 137(16): 2691-701, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20610481

RESUMEN

The development of neural crest cells involves an epithelial-mesenchymal transition (EMT) associated with the restriction of cadherin 6B expression to the pre-migratory neural crest cells (PMNCCs), as well as a loss of N-cadherin expression. We find that cadherin 6B, which is highly expressed in PMNCCs, persists in early migrating neural crest cells and is required for their emigration from the neural tube. Cadherin 6B-expressing PMNCCs exhibit a general loss of epithelial junctional polarity and acquire motile properties before their delamination from the neuroepithelium. Cadherin 6B selectively induces the de-epithelialization of PMNCCs, which is mediated by stimulation of BMP signaling, whereas N-cadherin inhibits de-epithelialization and BMP signaling. As BMP signaling also induces cadherin 6B expression and represses N-cadherin, cadherin-regulated BMP signaling may create two opposing feedback loops. Thus, the overall EMT of neural crest cells occurs via two distinct steps: a cadherin 6B and BMP signaling-mediated de-epithelialization, and a subsequent delamination through the basement membrane.


Asunto(s)
Proteínas Aviares/metabolismo , Cadherinas/metabolismo , Diferenciación Celular , Células Epiteliales/metabolismo , Cresta Neural/metabolismo , Transducción de Señal , Células Madre/metabolismo , Animales , Proteínas Aviares/genética , Proteínas Morfogenéticas Óseas/metabolismo , Cadherinas/genética , Movimiento Celular , Embrión de Pollo , Células Epiteliales/citología , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/citología , Células Madre/citología
17.
Cancers (Basel) ; 14(19)2022 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-36230633

RESUMEN

Breast tumor cells recruit bone marrow-derived mesenchymal stem cells (BM-MSCs) and alter their cellular characteristics to establish a tumor microenvironment. BM-MSCs enhance tumor angiogenesis through various mechanisms. We investigated the mechanisms by which BM-MSCs promote angiogenesis in response to breast tumor. Conditioned media from MDA-MB-231 (MDA CM) and MCF7 (MCF7 CM) breast tumor cells were used to mimic breast tumor conditions. An in vitro spheroid sprouting assay using human umbilical vein endothelial cells (HUVECs) was conducted to assess the angiogenesis-stimulating potential of BM-MSCs in response to breast tumors. The ROS inhibitor N-acetylcysteine (NAC) and JAK inhibitor ruxolitinib attenuated increased HIF-1α in BM-MSCs in response to MDA CM and MCF7 CM. HIF-1α knockdown or HIF-1ß only partially downregulated VEGF expression and, therefore, the sprouting capacity of HUVECs in response to conditioned media from BM-MSCs treated with MDA CM or MCF7 CM. Inactivation of the VEGF receptor using sorafenib completely inhibited the HUVECs' sprouting. Our results suggest that increased HIF-1α expression under normoxia in BM-MSCs in response to breast tumor cells is mediated by ROS and JAK/Stat3, and that both HIF-1α-dependent and -independent mechanisms increase VEGF expression in BM-MSCs to promote the angiogenic sprouting capacity of endothelial cells in a VEGF-dependent manner.

18.
Theranostics ; 11(14): 6786-6799, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093853

RESUMEN

Rationale: Bone marrow-derived mesenchymal stem cells (BM-MSCs) recruited into breast tumors regulate the behavior of tumor cells via various mechanisms and affect clinical outcomes. Although signaling molecules, such as transforming growth factor ß (TGF-ß), are known to transmit signals between BM-MSCs and breast tumor cells for recruiting BM-MSCs, it is unclear which specific intrinsic molecules involved in cell motility mediate the migration of BM-MSCs into breast tumor. It is also unclear as to how specific intrinsic molecules contribute to the migration. Methods: Conditioned medium (CM) from breast tumor cells (MCF-7 and MDA-MB-231) that simulates breast tumor secreting TGF-ß was used to examine the migration of BM-MSCs into breast tumors. A three-dimensional migration assay was performed to investigate the collective migration of BM-MSCs, maintaining cell-cell adhesion, toward breast tumor cells. Results: N-cadherin formed adherens junction-like structures on the intercellular borders of BM-MSCs, and TGF-ß increased the expression of N-cadherin on these borders. Knockdown of Smad4 impaired the TGF-ß-mediated increase in N-cadherin expression in BM-MSCs, but inhibitors of non-canonical TGF-ß pathways, such as extracellular signal-regulated kinases, Akt, and p38, did not affect it. siRNA-mediated knockdown of N-cadherin and Smad4 impaired the migration of BM-MSCs in response to TGF-ß. Conditioned medium from breast tumor cells also enhanced the expression of N-cadherin in BM-MSCs, but inactivation of TGF-ß type 1 receptor (TGFBR1) with SB505124 and TGFBR1 knockdown abolished the increase in N-cadherin expression. BM-MSCs collectively migrated toward CM from MDA-MB-231 in vitro while maintaining cell-cell adhesion through N-cadherin. Knockdown of N-cadherin abolished the migration of BM-MSCs toward the CM from breast tumor cells. Conclusion: In the present study, we identified N-cadherin, an intrinsic transmembrane molecule in adherens junction-like structures, on BM-MSCs as a mediator for the migration of these cells toward breast tumor. The expression of N-cadherin increases on the intercellular borders of BM-MSCs through the TGF-ß canonical signaling and they collectively migrate in response to breast tumor cells expressing TGF-ß via N-cadherin-dependent cell-cell adhesion. We, herein, introduce a novel promising strategy for controlling and re-engineering the breast tumor microenvironment.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/farmacología , Microambiente Tumoral/efectos de los fármacos , Antígenos CD/genética , Benzodioxoles/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Cadherinas/genética , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Humanos , Imidazoles/farmacología , Inmunohistoquímica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína Smad4/genética , Proteína Smad4/metabolismo , Microambiente Tumoral/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Biomedicines ; 9(11)2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34829800

RESUMEN

The prostate tumor microenvironment plays important roles in the metastasis and hormone-insensitive re-growth of tumor cells. Bone marrow-derived mesenchymal stem cells (BM-MSCs) are recruited into prostate tumors to facilitate tumor microenvironment formation. However, the specific intrinsic molecules mediating BM-MSCs' migration to prostate tumors are unknown. BM-MSCs' migration toward a conditioned medium (CM) of hormone-insensitive (PC3 and DU145) or hormone-sensitive (LNCaP) prostate tumor cells was investigated using a three-dimensional cell migration assay and a transwell migration assay. PC3 and DU145 expressed transforming growth factor-ß (TGF-ß), but LNCaP did not. Regardless of TGF-ß expression, BM-MSCs migrated toward the CM of PC3, DU145, or LNCaP. The CM of PC3 or DU145 expressing TGF-ß increased the phosphorylation of Smad2/3 in BM-MSCs. Inactivation of TGF-ß signaling in BM-MSCs using TGF-ß type 1 receptor (TGFBR1) inhibitors, SB505124, or SB431542 did not allow BM-MSCs to migrate toward the CM. The CM of PC3 or DU145 enhanced N-cadherin expression on BM-MSCs, but the LNCaP CM did not. SB505124, SB431542, and TGFBR1 knockdown prevented an increase in N-cadherin expression. N-cadherin knockdown inhibited the collective migration of BM-MSCs toward the PC3 CM. We identified N-cadherin as a mediator of BM-MSCs' migration toward hormone-insensitive prostate tumor cells expressing TGF-ß and introduced a novel strategy for controlling and re-engineering the prostate tumor microenvironment.

20.
Toxicol Res ; 37(4): 435-442, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34631500

RESUMEN

In vivo phototoxicity testing is important for predicting drug-induced phototoxicity in humans. Currently, there is no internationally validated in vivo test method for the photosafety evaluation of pharmaceuticals. In this study, we evaluated the phototoxicity of systemically administered drugs using SD rats. We first determined the appropriate ultraviolet A (UVA) dose using 8-methoxypsoralen, a well-known phototoxic drug. Compared to lower and higher UVA doses, we found that a UVA dose of 10 J/cm2 allowed for the detection of phototoxic responses in both a dose- and time-dependent manner. We next performed a phototoxicity study using seven pharmaceutical drugs which included known phototoxic and non-phototoxic drugs using a UVA dose of 10 J/cm2. In order to improve the accuracy of our assessment, we evaluated both gross skin findings as well as histopathological findings. Using gross skin findings alone resulted in an accuracy of 85.7% which could be increased to 100% accuracy when the gross skin findings were combined with histopathological findings. This study suggests that the inclusion of histopathological findings increases the accuracy of the phototoxicity evaluation of systemically administered drugs in SD rats. In conclusion, we found that for studying drug-induced phytotoxicity, a 10 J/cm2 UVA dose serves as the optimal radiation dose, and that the inclusion of histopathological findings increases the accuracy of the phototoxicity evaluation of the drugs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA