Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Semin Cancer Biol ; 92: 130-138, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37068553

RESUMEN

Tumor cells evolve in tumor microenvironment composed of multiple cell types. Among these, endothelial cells (ECs) are the major players in tumor angiogenesis, which is a driver of tumor progression and metastasis. Increasing evidence suggests that ECs also contribute to tumor progression and metastasis as they modify their phenotypes to differentiate into mesenchymal cells through a process known as endothelial-mesenchymal transition (EndoMT). This plasticity of ECs is mediated by various cytokines, including transforming growth factor-ß (TGF-ß), and modulated by other stimuli depending on the cellular contexts. Recent lines of evidence have shown that EndoMT is involved in various steps of tumor progression, including tumor angiogenesis, intravasation and extravasation of cancer cells, formation of cancer-associated fibroblasts, and cancer therapy resistance. In this review, we summarize current updates on EndoMT, highlight the roles of EndoMT in tumor progression and metastasis, and underline targeting EndoMT as a potential therapeutic strategy.


Asunto(s)
Células Endoteliales , Factor de Crecimiento Transformador beta , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Células Endoteliales/metabolismo , Microambiente Tumoral/genética , Endotelio , Citocinas/metabolismo , Neovascularización Patológica/metabolismo , Transición Epitelial-Mesenquimal/genética
2.
Cancer Sci ; 115(2): 490-506, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38111334

RESUMEN

Tumor progression and metastasis are regulated by endothelial cells undergoing endothelial-mesenchymal transition (EndoMT), a cellular differentiation process in which endothelial cells lose their properties and differentiate into mesenchymal cells. The cells undergoing EndoMT differentiate through a spectrum of intermediate phases, suggesting that some cells remain in a partial EndoMT state and exhibit an endothelial/mesenchymal phenotype. However, detailed analysis of partial EndoMT has been hampered by the lack of specific markers. Transforming growth factor-ß (TGF-ß) plays a central role in the induction of EndoMT. Here, we showed that inhibition of TGF-ß signaling suppressed EndoMT in a human oral cancer cell xenograft mouse model. By using genetic labeling of endothelial cell lineage, we also established a novel EndoMT reporter cell system, the EndoMT reporter endothelial cells (EMRECs), which allow visualization of sequential changes during TGF-ß-induced EndoMT. Using EMRECs, we characterized the gene profiles of multiple EndoMT stages and identified CD40 as a novel partial EndoMT-specific marker. CD40 expression was upregulated in the cells undergoing partial EndoMT, but decreased in the full EndoMT cells. Furthermore, single-cell RNA sequencing analysis of human tumors revealed that CD40 expression was enriched in the population of cells expressing both endothelial and mesenchymal cell markers. Moreover, decreased expression of CD40 in EMRECs enhanced TGF-ß-induced EndoMT, suggesting that CD40 expressed during partial EndoMT inhibits transition to full EndoMT. The present findings provide a better understanding of the mechanisms underlying TGF-ß-induced EndoMT and will facilitate the development of novel therapeutic strategies targeting EndoMT-driven cancer progression and metastasis.


Asunto(s)
Células Endoteliales , Transición Endotelial-Mesenquimatosa , Animales , Humanos , Ratones , Células Cultivadas , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/genética , Antígenos CD40/metabolismo
3.
Part Fibre Toxicol ; 19(1): 27, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35395797

RESUMEN

BACKGROUND: In Japan, six workers handling cross-linked water-soluble acrylic acid polymer (CWAAP) at a chemical plant suffered from lung diseases, including fibrosis, interstitial pneumonia, emphysema, and pneumothorax. We recently demonstrated that inhalation of CWAAP-A, one type of CWAAP, causes pulmonary disorders in rats. It is important to investigate dose-response relationships and recoverability from exposure to CWAAPs for establishing occupational health guidelines, such as setting threshold limit value for CWAAPs in the workplace. METHODS: Male and female F344 rats were exposed to 0.3, 1, 3, or 10 mg/m3 CWAAP-A for 6 h/day, 5 days/week for 13 weeks using a whole-body inhalation exposure system. At 1 h, 4 weeks, and 13 weeks after the last exposure the rats were euthanized and blood, bronchoalveolar lavage fluid, and all tissues including lungs and mediastinal lymph nodes were collected and subjected to biological and histopathological analyses. In a second experiment, male rats were pre-treated with clodronate liposome or polymorphonuclear leukocyte-neutralizing antibody to deplete macrophages or neutrophils, respectively, and exposed to CWAAP-A for 6 h/day for 2 days. RESULTS: CWAAP-A exposure damaged only the alveoli. The lowest observed adverse effect concentration (LOAEC) was 1 mg/m3 and the no observed adverse effect concentration (NOAEC) was 0.3 mg/m3. Rats of both sexes were able to recover from the tissue damage caused by 13 weeks exposure to 1 mg/m3 CWAAP-A. In contrast, tissue damage caused by exposure to 3 and 10 mg/m3 was irreversible due to the development of interstitial lung lesions. There was a gender difference in the recovery from CWAAP-A induced pulmonary disorders, with females recovering less than males. Finally, acute lung effects caused by CWAAP-A were significantly reduced by depletion of alveolar macrophages. CONCLUSIONS: Pulmonary damage caused by inhalation exposure to CWAAP-A was dose-dependent, specific to the lung and lymph nodes, and acute lung damage was ameliorated by depleting macrophages in the lungs. CWAAP-A had both a LOAEC and a NOAEC, and tissue damage caused by exposure to 1 mg/m3 CWAAP-A was reversible: recovery in female rats was less than for males. These findings indicate that concentration limits for CWAAPs in the workplace can be determined.


Asunto(s)
Exposición por Inhalación , Neumonía , Acrilatos , Animales , Líquido del Lavado Bronquioalveolar , Femenino , Exposición por Inhalación/efectos adversos , Pulmón , Masculino , Neumonía/patología , Polímeros/farmacología , Ratas , Ratas Endogámicas F344 , Agua
4.
J Biol Chem ; 295(36): 12559-12572, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32631954

RESUMEN

Tumor progression is governed by various growth factors and cytokines in the tumor microenvironment (TME). Among these, transforming growth factor-ß (TGF-ß) is secreted by various cell types residing in the TME and promotes tumor progression by inducing the epithelial-to-mesenchymal transition (EMT) of cancer cells and tumor angiogenesis. TGF-ß comprises three isoforms, TGF-ß1, -ß2, and -ß3, and transduces intracellular signals via TGF-ß type I receptor (TßRI) and TGF-ß type II receptor (TßRII). For the purpose of designing ligand traps that reduce oncogenic signaling in the TME, chimeric proteins comprising the ligand-interacting ectodomains of receptors fused with the Fc portion of immunoglobulin are often used. For example, chimeric soluble TßRII (TßRII-Fc) has been developed as an effective therapeutic strategy for targeting TGF-ß ligands, but several lines of evidence indicate that TßRII-Fc more effectively traps TGF-ß1 and TGF-ß3 than TGF-ß2, whose expression is elevated in multiple cancer types. In the present study, we developed a chimeric TGF-ß receptor containing both TßRI and TßRII (TßRI-TßRII-Fc) and found that TßRI-TßRII-Fc trapped all TGF-ß isoforms, leading to inhibition of both the TGF-ß signal and TGF-ß-induced EMT of oral cancer cells, whereas TßRII-Fc failed to trap TGF-ß2. Furthermore, we found that TßRI-TßRII-Fc suppresses tumor growth and angiogenesis more effectively than TßRII-Fc in a subcutaneous xenograft model of oral cancer cells with high TGF-ß expression. These results suggest that TßRI-TßRII-Fc may be a promising tool for targeting all TGF-ß isoforms in the TME.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Receptores Fc/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Carcinoma de Células Escamosas/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Boca/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Receptores Fc/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral
5.
Cancer Sci ; 112(4): 1633-1643, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33565179

RESUMEN

Tumor metastasis is the leading cause of death worldwide and involves an extremely complex process composed of multiple steps. Our previous study demonstrated that apoptosis signal-regulating kinase 1 (ASK1) deficiency in mice attenuates tumor metastasis in an experimental lung metastasis model. However, the steps of tumor metastasis regulated by ASK1 remain unclear. Here, we showed that ASK1 deficiency in mice promotes natural killer (NK) cell-mediated intravascular tumor cell clearance in the initial hours of metastasis. In response to tumor inoculation, ASK1 deficiency upregulated immune response-related genes, including interferon-gamma (IFNγ). We also revealed that NK cells are required for these anti-metastatic phenotypes. ASK1 deficiency augmented cytokine production chemoattractive to NK cells possibly through induction of the ligand for NKG2D, a key activating receptor of NK cells, leading to further recruitment of NK cells into the lung. These results indicate that ASK1 negatively regulates NK cell-dependent anti-tumor immunity and that ASK1-targeted therapy can provide a new tool for cancer immunotherapy to overcome tumor metastasis.


Asunto(s)
Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Metástasis de la Neoplasia/patología , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Inmunoterapia/métodos , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/inmunología , Células RAW 264.7
6.
Cancer Sci ; 111(8): 2954-2964, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32519375

RESUMEN

Cancer cachexia, characterized by continuous muscle wasting, is a key determinant of cancer-related death; however, there are few medical treatments to combat it. Myostatin (MSTN)/growth differentiation factor 8 (GDF-8), which is a member of the transforming growth factor-ß family, is secreted in an inactivated form noncovalently bound to the prodomain, negatively regulating the skeletal muscle mass. Therefore, inhibition of MSTN signaling is expected to serve as a therapeutic target for intractable muscle wasting diseases. Here, we evaluated the inhibitory effect of peptide-2, an inhibitory core of mouse MSTN prodomain, on MSTN signaling. Peptide-2 selectively suppressed the MSTN signal, although it had no effect on the activin signal. In contrast, peptide-2 slightly inhibited the GDF-11 signaling pathway, which is strongly related to the MSTN signaling pathway. Furthermore, we found that the i.m. injection of peptide-2 to tumor-implanted C57BL/6 mice alleviated muscle wasting in cancer cachexia. Although peptide-2 was unable to improve the loss of heart weight and fat mass when cancer cachexia model mice were injected with it, peptide-2 increased the gastrocnemius muscle weight and muscle cross-sectional area resulted in the enhanced grip strength in cancer cachexia mice. Consequently, the model mice treated with peptide-2 could survive longer than those that did not undergo this treatment. Our results suggest that peptide-2 might be a novel therapeutic candidate to suppress muscle wasting in cancer cachexia.


Asunto(s)
Caquexia/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/complicaciones , Miostatina/antagonistas & inhibidores , Neoplasias/complicaciones , Péptidos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Caquexia/etiología , Caquexia/patología , Factores de Diferenciación de Crecimiento/metabolismo , Células Hep G2 , Humanos , Masculino , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Miostatina/genética , Miostatina/metabolismo , Péptidos/genética , Péptidos/farmacología , Precursores de Proteínas/genética
7.
Cancer Sci ; 111(7): 2385-2399, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32385953

RESUMEN

The tumor microenvironment (TME) consists of various components including cancer cells, tumor vessels, cancer-associated fibroblasts (CAFs), and inflammatory cells. These components interact with each other via various cytokines, which often induce tumor progression. Thus, a greater understanding of TME networks is crucial for the development of novel cancer therapies. Many cancer types express high levels of TGF-ß, which induces endothelial-to-mesenchymal transition (EndMT), leading to formation of CAFs. Although we previously reported that CAFs derived from EndMT promoted tumor formation, the molecular mechanisms underlying these interactions remain to be elucidated. Furthermore, tumor-infiltrating inflammatory cells secrete various cytokines, including TNF-α. However, the role of TNF-α in TGF-ß-induced EndMT has not been fully elucidated. Therefore, this study examined the effect of TNF-α on TGF-ß-induced EndMT in human endothelial cells (ECs). Various types of human ECs underwent EndMT in response to TGF-ß and TNF-α, which was accompanied by increased and decreased expression of mesenchymal cell and EC markers, respectively. In addition, treatment of ECs with TGF-ß and TNF-α exhibited sustained activation of Smad2/3 signals, which was presumably induced by elevated expression of TGF-ß type I receptor, TGF-ß2, activin A, and integrin αv, suggesting that TNF-α enhanced TGF-ß-induced EndMT by augmenting TGF-ß family signals. Furthermore, oral squamous cell carcinoma-derived cells underwent epithelial-to-mesenchymal transition (EMT) in response to humoral factors produced by TGF-ß and TNF-α-cultured ECs. This EndMT-driven EMT was blocked by inhibiting the action of TGF-ßs. Collectively, our findings suggest that TNF-α enhances TGF-ß-dependent EndMT, which contributes to tumor progression.


Asunto(s)
Transición Epitelial-Mesenquimal , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Biomarcadores , Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular , Células Cultivadas , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , FN-kappa B/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Microambiente Tumoral/genética , Factor de Necrosis Tumoral alfa/farmacología
8.
Cancer Sci ; 108(1): 151-155, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28133920

RESUMEN

Clinical development of anti-angiogenic agents has been a major landmark in cancer therapy for several types of cancers. Signals mediated by both vascular endothelial growth factor (VEGF) and bone morphogenetic protein (BMP)-9 and 10 have been implicated in tumor angiogenesis. However, previous studies have shown that targeting the individual signals was not sufficiently effective in retarding tumor growth in certain preclinical and clinical conditions. In the present study, we developed a novel decoy chimeric receptor that traps both VEGF and BMP-9/10. Single targeting of either VEGF or BMP-9/10 signals significantly reduced the formation of tumor vessels in a mouse xenograft model of human pancreatic cancer; however, it did not show significant therapeutic effects on tumor growth. In contrast, dual targeting of the angiogenic signals resulted in more significant inhibition of tumor angiogenesis, leading to delay of tumor growth. Our findings suggest that simultaneous blockade of VEGF and BMP-9/10 signals is a promising therapeutic strategy for the cancers that are resistant to anti-VEGF and BMP-9/10 therapies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/farmacología , Receptores de Activinas Tipo II/uso terapéutico , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Femenino , Factor 2 de Diferenciación de Crecimiento/antagonistas & inhibidores , Factor 2 de Diferenciación de Crecimiento/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/irrigación sanguínea , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/química , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Sci ; 108(3): 419-426, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28064471

RESUMEN

Vasohibin-2 (VASH2) is a homolog of VASH1, an endothelium-derived angiogenesis inhibitor. Vasohibin-2 is mainly expressed in cancer cells, and has been implicated in the progression of cancer by inducing angiogenesis and tumor growth. Although VASH2 has been recently reported to be involved in epithelial-mesenchymal transition (EMT), its precise roles are obscure. The aim of the present study was to clarify the role of VASH2 in the EMT of cancer cells in relation to transforming growth factor-ß (TGF-ß) signaling, which is a major stimulator of EMT. Decreased expression of VASH2 in ovarian cancer cells significantly repressed the expression of TGF-ß type I receptor, namely activin receptor-like kinase 5. Transforming growth factor-ß1-induced phosphorylation of Smad2 and Smad3 was markedly decreased in VASH2 knockdown cells while the expression of Smad2 and Smad3 was unchanged. Accordingly, the responses to TGF-ß1 shown by promoter assay and plasminogen activator inhibitor type 1 expression were significantly attenuated in VASH2 knockdown cells. Furthermore, knockdown of VASH2 in cancer cells abrogated the TGF-ß1-induced reduced expression of epithelial markers including E-cadherin, and the elevated expression of mesenchymal markers including fibronectin, ZEB2, and Snail2, suggesting that endogenous VASH2 is required for TGF-ß1-induced EMT. In accordance with these results, the effects of TGF-ß1 on cell morphology, migration, invasion, and MMP2 expression were also abrogated when VASH2 was knocked down. These results indicate that VASH2 played a significant role in the EMT by modulating the TGF-ß signaling. We propose that VASH2 would be a novel molecular target for the prevention of EMT in cancers.


Asunto(s)
Proteínas Angiogénicas/genética , Transición Epitelial-Mesenquimal/genética , Neoplasias Ováricas/patología , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo , Cadherinas/biosíntesis , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Fibronectinas/biosíntesis , Proteínas de Homeodominio/biosíntesis , Humanos , Metaloproteinasa 2 de la Matriz/biosíntesis , Neovascularización Patológica/genética , Neoplasias Ováricas/genética , Fosforilación/genética , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Proteínas Represoras/biosíntesis , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factores de Transcripción de la Familia Snail/biosíntesis , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
11.
Proc Natl Acad Sci U S A ; 110(47): 18940-5, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24133138

RESUMEN

Lymphatic vessels (LVs) play critical roles in the maintenance of fluid homeostasis and in pathological conditions, including cancer metastasis. Although mutations in ALK1, a member of the transforming growth factor (TGF)-ß/bone morphogenetic protein (BMP) receptor family, have been linked to hereditary hemorrhagic telangiectasia, a human vascular disease, the roles of activin receptor-like kinase 1 (ALK-1) signals in LV formation largely remain to be elucidated. We show that ALK-1 signals inhibit LV formation, and LVs were enlarged in multiple organs in Alk1-depleted mice. These inhibitory effects of ALK-1 signaling were mediated by BMP-9, which decreased the number of cultured lymphatic endothelial cells. Bmp9-deficient mouse embryos consistently exhibited enlarged dermal LVs. BMP-9 also inhibited LV formation during inflammation and tumorigenesis. BMP-9 downregulated the expression of the transcription factor prospero-related homeobox 1, which is necessary to maintain lymphatic endothelial cell identity. Furthermore, silencing prospero-related homeobox 1 expression inhibited lymphatic endothelial cell proliferation. Our findings reveal a unique molecular basis for the physiological and pathological roles of BMP-9/ALK-1 signals in LV formation.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Factor 2 de Diferenciación de Crecimiento/metabolismo , Vasos Linfáticos/fisiología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Peritonitis/fisiopatología , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Cartilla de ADN/genética , Diafragma/patología , Perfilación de la Expresión Génica , Células HEK293 , Técnicas Histológicas , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Fluorescente , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Cancer Sci ; 105(9): 1116-23, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24981766

RESUMEN

The lymphatic system plays important roles not only in the physiological processes, such as maintenance of tissue fluid homeostasis, but also in pathological processes including the lymph node metastasis of tumor cells. Therefore, understanding of the molecular mechanisms underlying lymphatic vessel formation is crucial. Previous studies have shown that proliferation and migration of lymphatic endothelial cells (LECs) are activated by multiple types of signals mediated by tyrosine kinase receptors such as vascular endothelial growth factor receptor (VEGFR) 3. Although signals mediated by platelet-derived growth factor receptor ß (PDGFRß) have been implicated in lymphangiogenesis, the mechanisms explaining how PDGFRß expression is maintained in LECs remain to be fully elucidated. In the present study, we show that PDGFRß expression in LECs is maintained by Prox1 transcription factor. Knockdown of Prox1 expression in human dermal LECs decreased the expression of PDGFRß, leading to the lowered migration of human dermal LECs towards PDGF-BB. Furthermore, we found that PDGF signals play important roles in inflammatory lymphangiogenesis in a chronic aseptic peritonitis model. Intraperitoneal administration of imatinib, a potent inhibitor of PDGFRß, and transduction of PDGFRß/Fc chimeric protein by an adenoviral system both reduced inflammatory lymphangiogenesis induced by thioglycollate in mice. We also found that the expression of PDGFRß/Fc reduced tumor lymphangiogenesis in a BxPC3 human pancreatic cancer xenograft model. These findings suggest that PDGFRß is one of the key mediators of lymphatic vessel formation acting downstream of Prox1.


Asunto(s)
Proteínas de Homeodominio/fisiología , Vasos Linfáticos/fisiopatología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Supresoras de Tumor/fisiología , Animales , Antineoplásicos/farmacología , Becaplermina , Benzamidas/farmacología , Línea Celular Tumoral , Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Mesilato de Imatinib , Linfangiogénesis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-sis/fisiología , Pirimidinas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal
13.
J Cell Biol ; 223(3)2024 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-38353696

RESUMEN

The microtubule-associated protein MAP1B has been implicated in axonal growth and brain development. We found that MAP1B is highly expressed in the most aggressive and deadliest breast cancer subtype, triple-negative breast cancer (TNBC), but not in other subtypes. Expression of MAP1B was found to be highly correlated with poor prognosis. Depletion of MAP1B in TNBC cells impairs cell migration and invasion concomitant with a defect in tumorigenesis. We found that MAP1B interacts with key components for invadopodia formation, cortactin, and Tks5, the latter of which is a PtdIns(3,4)P2-binding and scaffold protein that localizes to invadopodia. We also found that Tks5 associates with microtubules and supports the association between MAP1B and α-tubulin. In accordance with their interaction, depletion of MAP1B leads to Tks5 destabilization, leading to its degradation via the autophagic pathway. Collectively, these findings suggest that MAP1B is a convergence point of the cytoskeleton to promote malignancy in TNBC and thereby a potential diagnostic and therapeutic target for TNBC.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular , Cortactina , Proteínas Asociadas a Microtúbulos , Neoplasias de la Mama Triple Negativas , Humanos , Carcinogénesis/genética , Transformación Celular Neoplásica , Cortactina/genética , Proteínas Asociadas a Microtúbulos/genética , Neoplasias de la Mama Triple Negativas/genética , Células MDA-MB-231 , Proteínas Adaptadoras del Transporte Vesicular/genética , Microtúbulos/metabolismo , Citoesqueleto/metabolismo , Femenino , Animales , Ratones , Ratones Endogámicos BALB C , Podosomas/metabolismo , Tubulina (Proteína)/metabolismo
14.
iScience ; 27(4): 109528, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38595797

RESUMEN

Diabetic neuropathy (DN) is a major complication of diabetes mellitus. Chondroitin sulfate (CS) is one of the most important extracellular matrix components and is known to interact with various diffusible factors; however, its role in DN pathology has not been examined. Therefore, we generated CSGalNAc-T1 knockout (T1KO) mice, in which CS levels were reduced. We demonstrated that diabetic T1KO mice were much more resistant to DN than diabetic wild-type (WT) mice. We also found that interactions between pericytes and vascular endothelial cells were more stable in T1KO mice. Among the RNA-seq results, we focused on the transforming growth factor ß signaling pathway and found that the phosphorylation of Smad2/3 was less upregulated in T1KO mice than in WT mice under hyperglycemic conditions. Taken together, a reduction in CS level attenuates DN progression, indicating that CS is an important factor in DN pathogenesis.

15.
J Cell Sci ; 124(Pt 16): 2753-62, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21807940

RESUMEN

Prox1 plays pivotal roles during embryonic lymphatic development and maintenance of adult lymphatic systems by modulating the expression of various lymphatic endothelial cell (LEC) markers, such as vascular endothelial growth factor receptor 3 (VEGFR3). However, the molecular mechanisms by which Prox1 transactivates its target genes remain largely unknown. Here, we identified Ets-2 as a candidate molecule that regulates the functions of Prox1. Whereas Ets-2 has been implicated in angiogenesis, its roles during lymphangiogenesis have not yet been elucidated. We found that endogenous Ets-2 interacts with Prox1 in LECs. Using an in vivo model of chronic aseptic peritonitis, we found that Ets-2 enhanced inflammatory lymphangiogenesis, whereas a dominant-negative mutant of Ets-1 suppressed it. Ets-2 also enhanced endothelial migration towards VEGF-C through induction of expression of VEGFR3 in collaboration with Prox1. Furthermore, we found that both Prox1 and Ets-2 bind to the VEGFR3 promoter in intact chromatin. These findings suggest that Ets family members function as transcriptional cofactors that enhance Prox1-induced lymphangiogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas de Homeodominio/metabolismo , Peritonitis/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Proteína Proto-Oncogénica c-ets-2/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Células Endoteliales/inmunología , Células Endoteliales/patología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Inflamación , Linfangiogénesis/genética , Linfangiogénesis/inmunología , Ratones , Ratones Endogámicos BALB C , Mutación/genética , Peritonitis/inducido químicamente , Peritonitis/genética , Peritonitis/fisiopatología , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-2/genética , ARN Interferente Pequeño/genética , Tioglicolatos/administración & dosificación , Proteínas Supresoras de Tumor/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Inflamm Regen ; 42(1): 9, 2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35130955

RESUMEN

Endothelial-mesenchymal transition (EndoMT), a cellular differentiation process in which endothelial cells (ECs) lose their properties and differentiate into mesenchymal cells, has been observed not only during development but also in various pathological states in adults, including cancer progression and organ/tissue fibrosis. Transforming growth factor-ß (TGF-ß), an inflammation-related cytokine, has been shown to play central roles in the induction of EndoMT. TGF-ß induces EndoMT by regulating the expression of various transcription factors, signaling molecules, and cellular components that confer ECs with mesenchymal characteristics. However, TGF-ß by itself is not necessarily sufficient to induce EndoMT to promote the progression of EndoMT-related diseases to a refractory extent. In addition to TGF-ß, additional activation by other inflammatory factors is often required to stabilize the progression of EndoMT. Since recent lines of evidence indicate that inflammatory signaling molecules act as enhancers of EndoMT, we summarize the roles of inflammatory factors in the induction of EndoMT and related diseases. We hope that this review will help to develop therapeutic strategies for EndoMT-related diseases by targeting inflammation-mediated EndoMT.

17.
Nat Commun ; 13(1): 5239, 2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-36097010

RESUMEN

The blood and lymphatic vasculature networks are not yet fully understood even in mouse because of the inherent limitations of imaging systems and quantification methods. This study aims to evaluate the usefulness of the tissue-clearing technology for visualizing blood and lymphatic vessels in adult mouse. Clear, unobstructed brain/body imaging cocktails and computational analysis (CUBIC) enables us to capture the high-resolution 3D images of organ- or area-specific vascular structures. To evaluate these 3D structural images, signals are first classified from the original captured images by machine learning at pixel base. Then, these classified target signals are subjected to topological data analysis and non-homogeneous Poisson process model to extract geometric features. Consequently, the structural difference of vasculatures is successfully evaluated in mouse disease models. In conclusion, this study demonstrates the utility of CUBIC for analysis of vascular structures and presents its feasibility as an analysis modality in combination with 3D images and mathematical frameworks.


Asunto(s)
Análisis de Datos , Vasos Linfáticos , Animales , Encéfalo/diagnóstico por imagen , Imagenología Tridimensional/métodos , Vasos Linfáticos/diagnóstico por imagen , Ratones , Tecnología
18.
Dev Cell ; 11(3): 301-12, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16950122

RESUMEN

To assess whether Smad signaling affects skin development, we generated transgenic mice in which a Smad antagonist, Smad7, was induced in keratinocytes, including epidermal stem cells. Smad7 transgene induction perturbed hair follicle morphogenesis and differentiation, but accelerated sebaceous gland morphogenesis. Further analysis revealed that independent of its role in anti-Smad signaling, Smad7 bound beta-catenin and induced beta-catenin degradation by recruiting an E3 ligase, Smurf2, to the Smad7/beta-catenin complex. Consequently, Wnt/beta-catenin signaling was suppressed in Smad7 transgenic hair follicles. Coexpression of the Smurf2 and Smad7 transgenes exacerbated Smad7-induced abnormalities in hair follicles and sebaceous glands. Conversely, when endogenous Smad7 was knocked down, keratinocytes exhibited increased beta-catenin protein and enhanced Wnt signaling. Our data reveal a mechanism for Smad7 in antagonizing Wnt/beta-catenin signaling, thereby shifting the skin differentiation program from forming hair follicles to sebaceous glands.


Asunto(s)
Epidermis/patología , Folículo Piloso/fisiología , Proteína smad7/fisiología , Células Madre/fisiología , beta Catenina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Regulación hacia Abajo , Epidermis/metabolismo , Folículo Piloso/metabolismo , Humanos , Queratinocitos/metabolismo , Ratones , Ratones Transgénicos , Morfogénesis , Glándulas Sebáceas/fisiología , Transducción de Señal , Proteína smad7/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Wnt/metabolismo
19.
J Cell Sci ; 122(Pt 21): 3923-30, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19825936

RESUMEN

During lymphatic development, Prox1 plays central roles in the differentiation of blood vascular endothelial cells (BECs) into lymphatic endothelial cells (LECs), and subsequently in the maturation and maintenance of lymphatic vessels. However, the molecular mechanisms by which Prox1 elicits these functions remain to be elucidated. Here, we identified FoxC2 and angiopoietin-2 (Ang2), which play important roles in the maturation of lymphatic vessels, as novel targets of Prox1 in mouse embryonic-stem-cell-derived endothelial cells (MESECs). Furthermore, we found that expression of HoxD8 was significantly induced by Prox1 in MESECs, a finding confirmed in human umbilical vein endothelial cells (HUVECs) and human dermal LECs (HDLECs). In mouse embryos, HoxD8 expression was significantly higher in LECs than in BECs. In a model of inflammatory lymphangiogenesis, diameters of lymphatic vessels of the diaphragm were increased by adenovirally transduced HoxD8. We also found that HoxD8 induces Ang2 expression in HDLECs and HUVECs. Moreover, we found that HoxD8 induces Prox1 expression in HUVECs and that knockdown of HoxD8 reduces this expression in HDLECs, suggesting that Prox1 expression in LECs is maintained by HoxD8. These findings indicate that transcriptional networks of Prox1 and HoxD8 play important roles in the maturation and maintenance of lymphatic vessels.


Asunto(s)
Angiopoyetina 2/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/metabolismo , Linfangiogénesis , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Angiopoyetina 2/genética , Animales , Células Cultivadas , Células Madre Embrionarias/metabolismo , Endotelio Linfático/citología , Endotelio Linfático/embriología , Endotelio Linfático/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Factores de Transcripción Forkhead/genética , Proteínas de Homeodominio/genética , Humanos , Vasos Linfáticos/citología , Vasos Linfáticos/embriología , Vasos Linfáticos/metabolismo , Ratones , Ratones Endogámicos BALB C , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
20.
Sci Rep ; 11(1): 17989, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34504254

RESUMEN

Prevascularized artificial three-dimensional (3D) tissues are effective biomaterials for regenerative medicine. We have previously established a scaffold-free 3D artificial vascular tissue from normal human dermal fibroblasts (NHDFs) and umbilical vein-derived endothelial cells (HUVECs) by layer-by-layer cell coating technique. In this study, we constructed an artificial vascular tissue constructed by human adipose tissue-derived stromal cells (hASCs) and HUVECs (ASCVT) by a modified technique with cryopreservation. ASCVT showed a higher thickness with more dense vascular networks than the 3D tissue based on NHDFs. Correspondingly, 3D-cultured ASCs showed higher expression of several angiogenesis-related factors, including vascular endothelial growth factor-A and hepatic growth factor, compared to that of NHDFs. Moreover, perivascular cells in ASCVT were detected by pericyte markers, suggesting the differentiation of hASCs into pericyte-like cells. Subcutaneous transplantation of ASCVTs to nude mice resulted in an engraftment with anastomosis of host's vascular structures at 2 weeks after operation. In the engrafted tissue, the vascular network was surrounded by mural-like structure-forming hASCs, in which some parts developed to form vein-like structures at 4 weeks, suggesting the generation of functional vessel networks. These results demonstrated that cryopreserved human cells, including hASCs, could be used directly to construct the artificial transplantable tissue for regenerative medicine.


Asunto(s)
Criopreservación/métodos , Células Endoteliales de la Vena Umbilical Humana , Células Madre Mesenquimatosas , Ingeniería de Tejidos/métodos , Trasplantes/citología , Animales , Células Cultivadas , Femenino , Fibroblastos , Proteínas Fluorescentes Verdes/genética , Humanos , Recién Nacido , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Animales , Medicina Regenerativa/métodos , Andamios del Tejido , Trasplante de Tejidos/métodos , Transfección , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA