Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Cancer Sci ; 113(10): 3547-3557, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35849084

RESUMEN

Cancer cachexia is a multifactorial disease that causes continuous skeletal muscle wasting. Thereby, it seems to be a key determinant of cancer-related death. Although anamorelin, a ghrelin receptor agonist, has been approved in Japan for the treatment of cachexia, few medical treatments for cancer cachexia are currently available. Myostatin (MSTN)/growth differentiation factor 8, which belongs to the transforming growth factor-ß family, is a negative regulator of skeletal muscle mass, and inhibition of MSTN signaling is expected to be a therapeutic target for muscle-wasting diseases. Indeed, we have reported that peptide-2, an MSTN-inhibiting peptide from the MSTN prodomain, alleviates muscle wasting due to cancer cachexia. Herein, we evaluated the therapeutic benefit of myostatin inhibitory D-peptide-35 (MID-35), whose stability and activity were more improved than those of peptide-2 in cancer cachexia model mice. The biologic effects of MID-35 were better than those of peptide-2. Intramuscular administration of MID-35 effectively alleviated skeletal muscle atrophy in cachexia model mice, and the combination therapy of MID-35 with anamorelin increased food intake and maximized grip strength, resulting in longer survival. Our results suggest that this combination might be a novel therapeutic tool to suppress muscle wasting in cancer cachexia.


Asunto(s)
Productos Biológicos , Neoplasias , Animales , Productos Biológicos/farmacología , Caquexia/tratamiento farmacológico , Caquexia/etiología , Caquexia/patología , Modelos Animales de Enfermedad , Hidrazinas , Ratones , Músculo Esquelético , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/etiología , Atrofia Muscular/patología , Miostatina , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Oligopéptidos , Péptidos/farmacología , Receptores de Ghrelina/uso terapéutico , Factores de Crecimiento Transformadores/farmacología , Factores de Crecimiento Transformadores/uso terapéutico
3.
Biochem Biophys Res Commun ; 618: 79-85, 2022 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-35716599

RESUMEN

Arteriosclerosis is intimately associated with cardiovascular diseases. Recently, evidence accumulated that infection with Helicobacter pylori cagA-positive strains, which causes gastritis, peptic ulceration, and gastric cancer, is also involved in the development of arteriosclerosis. The cagA-encoded CagA protein is injected into the attached gastric epithelial cells via the type IV secretion system. We previously showed that CagA-containing exosomes are secreted from CagA-injected gastric epithelial cells and enter the systemic blood circulation, delivering CagA into endothelial cells. In the present study, transgenic mice were established in which CagA was selectively expressed in endothelial cells by Cre-loxP system. Treatment of the mice with a high-fat diet revealed that atherogenic lesions were induced in mice expressing CagA in vascular endothelial cells but not in CagA-nonexpressing mice. To investigate the effects of CagA on endothelial cells, we also established conditional CagA-expressing human vascular endothelial cells using the Tet-on system. Upon induction of CagA, a dramatic change in cell morphology was observed that was concomitantly associated with the loss of the endothelial cells to form tube-like structures. Induction of CagA also activated the pro-inflammatory transcription factor STAT3. Thus, exosome-delivered CagA deregulates signals that activates STAT3 in endothelial cells, which accelerates inflammation that promotes arteriosclerosis/atherosclerosis.


Asunto(s)
Arteriosclerosis , Infecciones por Helicobacter , Helicobacter pylori , Animales , Antígenos Bacterianos/metabolismo , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Proteínas Bacterianas/metabolismo , Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Ratones
4.
J Biol Chem ; 295(27): 9105-9120, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32371398

RESUMEN

Modification of the transforming growth factor ß (TGF-ß) signaling components by (de)ubiquitination is emerging as a key regulatory mechanism that controls cell signaling responses in health and disease. Here, we show that the deubiquitinating enzyme UBH-1 in Caenorhabditis elegans and its human homolog, ubiquitin C-terminal hydrolase-L1 (UCH-L1), stimulate DAF-7/TGF-ß signaling, suggesting that this mode of regulation of TGF-ß signaling is conserved across animal species. The dauer larva-constitutive C. elegans phenotype caused by defective DAF-7/TGF-ß signaling was enhanced and suppressed, respectively, by ubh-1 deletion and overexpression in the loss-of-function genetic backgrounds of daf7, daf-1/TGF-ßRI, and daf4/R-SMAD, but not of daf-8/R-SMAD. This suggested that UBH-1 may stimulate DAF-7/TGF-ß signaling via DAF-8/R-SMAD. Therefore, we investigated the effect of UCH-L1 on TGF-ß signaling via its intracellular effectors, i.e. SMAD2 and SMAD3, in mammalian cells. Overexpression of UCH-L1, but not of UCH-L3 (the other human homolog of UBH1) or of the catalytic mutant UCH-L1C90A, enhanced TGF-ß/SMAD-induced transcriptional activity, indicating that the deubiquitination activity of UCH-L1 is indispensable for enhancing TGF-ß/SMAD signaling. We also found that UCH-L1 interacts, deubiquitinates, and stabilizes SMAD2 and SMAD3. Under hypoxia, UCH-L1 expression increased and TGF-ß/SMAD signaling was potentiated in the A549 human lung adenocarcinoma cell line. Notably, UCH-L1-deficient A549 cells were impaired in tumorigenesis, and, unlike WT UCH-L1, a UCH-L1 variant lacking deubiquitinating activity was unable to restore tumorigenesis in these cells. These results indicate that UCH-L1 activity supports DAF-7/TGF-ß signaling and suggest that UCH-L1's deubiquitination activity is a potential therapeutic target for managing lung cancer.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Carcinogénesis/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Caenorhabditis elegans , Transformación Celular Neoplásica , Enzimas Desubicuitinizantes , Larva/metabolismo , Pulmón/metabolismo , Transducción de Señal/genética , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Ubiquitina Tiolesterasa/fisiología , Ubiquitinación
5.
J Biol Chem ; 294(13): 4966-4980, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30718277

RESUMEN

Transforming growth factor (TGF)-ß signaling in humans is stringently regulated to prevent excessive TGF-ß signaling. In tumors, TGF-ß signaling can both negatively and positively regulate tumorigenesis dependent on tumor type, but the reason for these opposite effects is unclear. TGF-ß signaling is mainly mediated via the Smad-dependent pathway, and herein we found that PDZK1-interacting protein 1 (PDZK1IP1) interacts with Smad4. PDZK1IP1 inhibited both the TGF-ß and the bone morphogenetic protein (BMP) pathways without affecting receptor-regulated Smad (R-Smad) phosphorylation. Rather than targeting R-Smad phosphorylation, PDZK1IP1 could interfere with TGF-ß- and BMP-induced R-Smad/Smad4 complex formation. Of note, PDZK1IP1 retained Smad4 in the cytoplasm of TGF-ß-stimulated cells. To pinpoint PDZK1IP1's functional domain, we created several PDZK1IP1 variants and found that its middle region, from Phe40 to Ala49, plays a key role in its Smad4-regulating activity. PDZK1IP1 knockdown enhanced the expression of the TGF-ß target genes Smad7 and prostate transmembrane protein androgen-induced (TMEPAI) upon TGF-ß stimulation. In contrast, PDZK1IP1 overexpression suppressed TGF-ß-induced reporter activities, cell migration, and cell growth inhibition. In a xenograft tumor model in which TGF-ß was previously shown to elicit tumor-promoting effects, PDZK1IP1 gain of function decreased tumor size and increased survival rates. Taken together, these findings indicate that PDZK1IP1 interacts with Smad4 and thereby suppresses the TGF-ß signaling pathway.


Asunto(s)
Proteínas de la Membrana/metabolismo , Neoplasias/metabolismo , Mapas de Interacción de Proteínas , Transducción de Señal , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Masculino , Ratones Endogámicos BALB C , Fosforilación
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.
J Biol Chem ; 292(10): 4099-4112, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28115518

RESUMEN

The intensity and duration of TGF-ß signaling determine the cellular biological response. How this is negatively regulated is not well understood. Here, we identified a novel negative regulator of TGF-ß signaling, transmembrane p24-trafficking protein 10 (TMED10). TMED10 disrupts the complex formation between TGF-ß type I (also termed ALK5) and type II receptors (TßRII). Misexpression studies revealed that TMED10 attenuated TGF-ß-mediated signaling. A 20-amino acid-long region from Thr91 to Glu110 within the extracellular region of TMED10 was found to be crucial for TMED10 interaction with both ALK5 and TßRII. Synthetic peptides corresponding to this region inhibit both TGF-ß-induced Smad2 phosphorylation and Smad-dependent transcriptional reporter activity. In a xenograft cancer model, where previously TGF-ß was shown to elicit tumor-promoting effects, gain-of-function and loss-of-function studies for TMED10 revealed a decrease and increase in the tumor size, respectively. Thus, we determined herein that TMED10 expression levels are the key determinant for efficiency of TGF-ß receptor complex formation and signaling.


Asunto(s)
Neoplasias Mamarias Animales/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Células Cultivadas , Femenino , Células HEK293 , Humanos , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta/genética , Proteínas de Transporte Vesicular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Cell ; 37(1): 123-34, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20129061

RESUMEN

Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine of key importance for controlling embryogenesis and tissue homeostasis. How TGF-beta signals are attenuated and terminated is not well understood. Here, we show that TMEPAI, a direct target gene of TGF-beta signaling, antagonizes TGF-beta signaling by interfering with TGF-beta type I receptor (TbetaRI)-induced R-Smad phosphorylation. TMEPAI can directly interact with R-Smads via a Smad interaction motif. TMEPAI competes with Smad anchor for receptor activation for R-Smad binding, thereby sequestering R-Smads from TbetaRI kinase activation. In mammalian cells, ectopic expression of TMEPAI inhibited TGF-beta-dependent regulation of plasminogen activator inhibitor-1, JunB, cyclin-dependent kinase inhibitors, and c-myc expression, whereas specific knockdown of TMEPAI expression prolonged duration of TGF-beta-induced Smad2 and Smad3 phosphorylation and concomitantly potentiated cellular responsiveness to TGF-beta. Consistently, TMEPAI inhibits activin-mediated mesoderm formation in Xenopus embryos. Therefore, TMEPAI participates in a negative feedback loop to control the duration and intensity of TGF-beta/Smad signaling.


Asunto(s)
Proteínas de la Membrana/fisiología , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activinas/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Mesodermo/crecimiento & desarrollo , Ratones , Modelos Biológicos , Células 3T3 NIH , ARN Mensajero/metabolismo , Xenopus
9.
Semin Cell Dev Biol ; 32: 98-106, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24910449

RESUMEN

Members of the transforming growth factor-ß (TGF-ß) family have been implicated in embryogenesis as well as in the determination of the cell fates of mouse and human embryonic stem (ES) cells, which are characterized by their self-renewal and pluripotency. The cellular responses to TGF-ß family signals are divergent depending on the cellular context and local environment. TGF-ß family signals play critical roles both in the maintenance of the pluripotent state of ES cells by inducing the expression of Nanog, Oct4, and Sox2, and in their differentiation into various cell types by regulating the expression of master regulatory genes. Moreover, multiple lines of evidence have suggested the importance of TGF-ß family signals in establishing induced pluripotent stem (iPS) cells. Since ES and iPS cells have great potential for applications in regenerative medicine, it is critical to figure out the mechanisms underlying their self-renewal, pluripotency, and differentiation. Here, we discuss the roles of TGF-ß family ligands and their downstream signaling molecules, Smad proteins, in the maintenance of the pluripotency and lineage specification of mouse and human ES and iPS cells.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Células Madre Pluripotentes/citología , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética
10.
Reprod Med Biol ; 15(4): 261-265, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-29259443

RESUMEN

McCune-Albright syndrome (MAS) is characterized by fibrous dysplasia (FD) of bone, café-au-lait skin pigmentation, and precocious puberty. Here we report a case of a 12-year-old girl with MAS presenting sexual precocity as initial signs, followed by FD of bone with her growth. She was referred to our hospital because of breast budding and abnormal genital bleeding at the age of 2.8 years. On physical examination, her height and weight were greater than two standard deviations of the mean ranges. Hormonal analysis revealed an elevated serum estradiol and suppressed luteinizing hormone and follicle-stimulating hormone production. Her bone age had advanced, and a 16-mm monocystic lesion was observed on her right ovary by pelvic ultrasonography. Considering the clinical and paraclinical findings, precocious pseudopuberty was suspected and periodic observations were started. Her estrogen "flare up" was transient and she had repeated similar episodes three times in the following 7 years. She complained of pain in her right hip at the age of 9.6 years, which was diagnosed as FD of bone by fluorodeoxyglucose-positron emission tomography. Although no café-au-lait skin pigmentation was observed, we made a preliminary diagnosis of MAS. Because clinical evidence for MAS can appear later in the course of recurrent autonomous cysts, careful observation and periodical assessments of patients with suspected MAS is necessary.

11.
J Biol Chem ; 289(18): 12680-92, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24627487

RESUMEN

Transforming growth factor (TGF)-ß signaling is deliberately regulated at multiple steps in its pathway from the extracellular microenvironment to the nucleus. However, how TGF-ß signaling is activated or attenuated is not fully understood. We recently identified transmembrane prostate androgen-induced RNA (TMEPAI), which is involved in a negative feedback loop of TGF-ß signaling. When we searched for a family molecule(s) for TMEPAI, we found C18ORF1, which, like TMEPAI, possesses two PY motifs and one Smad-interacting motif (SIM) domain. As expected, C18ORF1 could block TGF-ß signaling but not bone morphogenetic protein signaling. C18ORF1 bound to Smad2/3 via its SIM and competed with the Smad anchor for receptor activation for Smad2/3 binding to attenuate recruitment of Smad2/3 to the TGF-ß type I receptor (also termed activin receptor-like kinase 5 (ALK5)), in a similar fashion to TMEPAI. Knockdown of C18ORF1 prolonged duration of TGF-ß-induced Smad2 phosphorylation and concomitantly potentiated the expression of JunB, p21, and TMEPAI mRNAs induced by TGF-ß. Consistently, TGF-ß-induced cell migration was enhanced by the knockdown of C18ORF1. These results indicate that the inhibitory function of C18ORF1 on TGF-ß signaling is similar to that of TMEPAI. However, in contrast to TMEPAI, C18ORF1 was not induced upon TGF-ß signaling. Thus, we defined C18ORF1 as a surveillant of steady state TGF-ß signaling, whereas TMEPAI might help C18ORF1 to inhibit TGF-ß signaling in a coordinated manner when cells are stimulated with high levels of TGF-ß.


Asunto(s)
Proteínas de la Membrana/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Secuencias de Aminoácidos/genética , Animales , Sitios de Unión/genética , Western Blotting , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Proteínas de la Membrana/genética , Mutación , Fosforilación/efectos de los fármacos , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología
12.
Cancer Sci ; 106(11): 1524-33, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26296946

RESUMEN

Transforming growth factor-ß (TGF-ß) is a potent growth inhibitor in normal epithelial cells. However, a number of malignant tumors produce excessive amounts of TGF-ß, which affects the tumor-associated microenvironment by furthering the progression of tumorigenicity. Although it is known that the tumor-associated microenvironment often becomes hypoxic, how hypoxia influences TGF-ß signaling in this microenvironment is unknown. We investigated whether TGF-ß signaling is influenced by long-term exposure to hypoxia in Lewis lung carcinoma (LLC) cells. When the cells were exposed to hypoxia for more than 10 days, their morphology was remarkably changed to a spindle shape, and TGF-ß-induced Smad2 phosphorylation was enhanced. Concomitantly, TGF-ß-induced transcriptional activity was augmented under hypoxia, although TGF-ß did not influence the activity of a hypoxia-responsive reporter. Consistently, hypoxia influenced the expression of several TGF-ß target genes. Interestingly, the expressions of TGF-ß type I receptor (TßRI), also termed activin receptor like kinase-5 (ALK5), and TGF-ß1 were increased under the hypoxic condition. When we monitored the hypoxia-inducible factor-1 (HIF-1) transcriptional activity by use of green fluorescent protein governed by the hypoxia-responsive element in LLC cells transplanted into mice, TGF-ß-induced Smad2 phosphorylation was upregulated in vivo. Our results demonstrate that long-term exposure to hypoxia might alter responsiveness to TGF-ß signaling and affected the malignancy of LLC cells.


Asunto(s)
Carcinoma Pulmonar de Lewis/patología , Hipoxia de la Célula/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/fisiología , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Blood ; 119(22): 5320-8, 2012 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-22498737

RESUMEN

Transforming growth factor-ß (TGF-ß) is involved in vascular formation through activin receptor-like kinase (ALK)1 and ALK5. ALK5, which is expressed ubiquitously, phosphorylates Smad2 and Smad3, whereas endothelial cell (EC)-specific ALK1 activates Smad1 and Smad5. Because ALK5 kinase activity is required for ALK1 to transduce TGF-ß signaling via Smad1/5 in ECs, ALK5 knockout (KO) mice were not able to give us the precise mechanisms by which TGF-ß/ALK5/Smad2/3 signaling is implicated in angiogenesis. To delineate the role of Smad2/3 signaling in endothelium, the Smad2 gene in Smad3 KO mice was selectively deleted in ECs using Tie2-Cre transgenic mice, termed EC-specific Smad2/3 double KO (EC-Smad2/3KO) mice. EC-Smad2/3KO embryos revealed hemorrhage leading to embryonic lethality around E12.5. EC-Smad2/3KO embryos exhibited no abnormality of vasculogenesis and angiogenesis in both the yolk sac and the whole embryo, whereas vascular maturation was incomplete because of inadequate assembly of mural cells in the vasculature. Wide gaps between ECs and mural cells could be observed in the vasculature of EC-Smad2/3KO mice because of reduced expression of N-cadherin and sphingosine-1-phosphate receptor-1 (S1PR1) in ECs from those mice. These results indicated that Smad2/3 signaling in ECs is indispensable for maintenance of vascular integrity via the fine-tuning of N-cadherin, VE-cadherin, and S1PR1 expressions in the vasculature.


Asunto(s)
Cadherinas/biosíntesis , Endotelio/metabolismo , Regulación de la Expresión Génica/fisiología , Receptores de Lisoesfingolípidos/biosíntesis , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo II , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Cadherinas/genética , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Lisoesfingolípidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Proteína Smad2/genética , Proteína smad3/genética , Receptores de Esfingosina-1-Fosfato , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
14.
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
15.
Blood ; 115(20): 4138-47, 2010 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-20231428

RESUMEN

E2-2 belongs to the basic helix-loop-helix (bHLH) family of transcription factors. E2-2 associates with inhibitor of DNA binding (Id) 1, which is involved in angiogenesis. In this paper, we demonstrate that E2-2 interacts with Id1 and provide evidence that this interaction potentiates angiogenesis. Mutational analysis revealed that the HLH domain of E2-2 is required for the interaction with Id1 and vice versa. In addition, Id1 interfered with E2-2-mediated effects on luciferase reporter activities. Interestingly, injection of E2-2-expressing adenoviruses into Matrigel plugs implanted under the skin blocked in vivo angiogenesis. In contrast, the injection of Id1-expressing adenoviruses rescued E2-2-mediated inhibition of in vivo angiogenic reaction. Consistent with the results of the Matrigel plug assay, E2-2 could inhibit endothelial cell (EC) migration, network formation, and proliferation. On the other hand, knockdown of E2-2 in ECs increased EC migration. The blockade of EC migration by E2-2 was relieved by exogenous expression of Id1. We also demonstrated that E2-2 can perturb VEGFR2 expression via inhibition of VEGFR2 promoter activity. This study suggests that E2-2 can maintain EC quiescence and that Id1 can counter this effect.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Endotelio Vascular/fisiología , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Neovascularización Patológica/prevención & control , Factores de Transcripción TCF/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Western Blotting , Células COS , Células Cultivadas , Chlorocebus aethiops , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Inmunoprecipitación , Proteína 1 Inhibidora de la Diferenciación/genética , Ratones , Neovascularización Patológica/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción TCF/antagonistas & inhibidores , Factores de Transcripción TCF/genética , Factor de Transcripción 4 , Proteína 2 Similar al Factor de Transcripción 7 , Transcripción Genética , Técnicas del Sistema de Dos Híbridos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Sensors (Basel) ; 12(4): 4974-85, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666071

RESUMEN

Cardiovascular disease is the leading cause of death worldwide, with high medical costs and rates of disability. It is therefore important to evaluate the use of cardiovascular biomarkers in the early diagnosis of coronary artery disease (CAD). We have screened a variety of recently identified bioactive peptides candidates in anticipation that they would allow detection of atherosclerotic CAD. Especially, we have focused on novel anti-atherogenic peptides as indicators and negative risk factors for CAD. In vitro, in vivo and clinical studies indicated that human adiponectin, heregulin-ß(1), glucagon-like peptide-1 (GLP-1), and salusin-α, peptides of 244, 71, 30, and 28 amino acids, respectively, attenuate the development and progression of atherosclerotic lesions by suppressing macrophage foam cell formation via down-regulation of acyl-coenzyme A: cholesterol acyltransferase-1. Circulating levels of these peptides in the blood are significantly decreased in patients with CAD compared to patients without CAD. Receiver operating characteristic analyses showed that salusin-α is a more useful biomarker, with better sensitivity and specificity, compared with the others for detecting CAD. Therefore, salusin-α, heregulin-ß(1), adiponectin, and/or GLP-1, alone or in various combinations, may be useful as biomarkers for atherosclerotic CAD.


Asunto(s)
Aterosclerosis/metabolismo , Biomarcadores/metabolismo , Enfermedad Coronaria/metabolismo , Péptidos/metabolismo , Humanos
17.
Front Physiol ; 13: 1081376, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36589453

RESUMEN

Transforming growth factor (TGF)-ß and its family members, including bone morphogenetic proteins (BMPs), nodal proteins, and activins, are implicated in the development and maintenance of various organs. Here, we review its role in the lymphatic vascular system (the secondary vascular system in vertebrates), which plays a crucial role in various physiological and pathological processes, participating in the maintenance of the normal tissue fluid balance, immune cell trafficking, and fatty acid absorption in the gut. The lymphatic system is associated with pathogenesis in multiple diseases, including lymphedema, inflammatory diseases, and tumor metastasis. Lymphatic vessels are composed of lymphatic endothelial cells, which differentiate from blood vascular endothelial cells (BECs). Although TGF-ß family signaling is essential for maintaining blood vessel function, little is known about the role of TGF-ß in lymphatic homeostasis. Recently, we reported that endothelial-specific depletion of TGF-ß signaling affects lymphatic function. These reports suggest that TGF-ß signaling in lymphatic endothelial cells maintains the structure of lymphatic vessels and lymphatic homeostasis, and promotes tumor lymphatic metastasis. Suppression of TGF-ß signaling in lymphatic endothelial cells may therefore be effective in inhibiting cancer metastasis. We highlight recent advances in understanding the roles of TGF-ß signaling in the formation and maintenance of the lymphatic system.

18.
J Biol Chem ; 285(49): 38023-33, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-20889500

RESUMEN

The TGF-ß and Wnt pathways are involved in cell fate and tumorigenicity. A recent report indicated that a TGF-ß target gene, TMEPAI (transmembrane prostate androgen-induced RNA), is possibly also a downstream target of Wnt signaling. Although TMEPAI was believed to be involved in tumorigenicity because of its blockage of TGF-ß signaling, how TGF-ß and Wnt signals affect the activation of the TMEPAI gene is not well understood. Herein, we show that the TMEPAI promoter is regulated synergistically by TGF-ß/Smad and Wnt/ß-catenin/T cell factor (TCF) 7L2. The critical cis-element for dual signals, termed TGF-ß-responsive TCF7L2-binding element (TTE), is located in intron 1 of the TMEPAI gene. TCF7L2, but not Smad proteins, bound to TTE, whereas the disruption of TTE by mutagenesis remarkably counteracted both TGF-ß and TCF7L2 responses. The introduction of mutations in critical Smad-binding elements blocked the activation of the TMEPAI promoter by TCF7L2. Furthermore, our DNA-protein interaction experiments revealed the indirect binding of TCF7L2 to Smad-binding elements via Smad3 upon TGF-ß stimulation as well as its TGF-ß-dependent association with TTE. We demonstrate that the Wnt/ß-catenin/TCF7L2 pathway is preferentially able to alter the transcriptional regulation of the TGF-ß-target gene, TMEPAI.


Asunto(s)
Proteínas de la Membrana/biosíntesis , Elementos de Respuesta/fisiología , Transducción de Señal/fisiología , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Transcripción Genética/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células COS , Chlorocebus aethiops , Humanos , Proteínas de la Membrana/genética , Ratones , Mutación , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/genética , Factor de Crecimiento Transformador beta/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
19.
Growth Factors ; 29(5): 163-73, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21913798

RESUMEN

Transforming growth factor-ß (TGF-ß) family signaling regulates cell growth and differentiation of many different cell types and is widely involved in the regulation of homeostasis during both embryogenesis and adult life. Therefore, aberrant TGF-ß family signal transduction is linked to congenital disorders, tumorigenicity, and fibrosis, which can be life-threatening. A specific receptor-ligand complex initiates transduction of TGF-ß family signaling to the nucleus via intracellular signal molecules, mainly Smads, whereby a number of bioactivities such as wound healing, immunomodulation, apoptosis, and angiogenesis are controlled. To avoid an excess of TGF-ß family signaling in cells, the duration and intensity of the TGF-ß family signal appear to be subject to elaborate regulation. In this paper, we describe recent advances in the understanding of how TGF-ß family signals are perturbed and terminated to maintain homeostasis in cells.


Asunto(s)
Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Apoptosis/fisiología , Diferenciación Celular , Aumento de la Célula , Proliferación Celular , Desarrollo Embrionario , Humanos , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Neoplasias/fisiopatología , Neovascularización Fisiológica , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/genética
20.
Cancer Sci ; 102(10): 1808-14, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21722264

RESUMEN

The basic helix­loop­helix protein E2-2 is known to play a role in quiescence of endothelial cells (ECs). However, it is unclear how the activity of E2-2 is controlled in the cells. In this study, we identified FAM96B as an interaction partner of E2-2. FAM96B interfered with E2-2-mediated effects on luciferase reporter activities. Furthermore, the suppression of vascular endothelial growth factor receptor 2 promoter activity by E2-2 was rescued by the expression of FAM96B in a dose-dependent manner. Interestingly, FAM96B decreased the expression of ectopic and endogenous E2-2 proteins. Mutational analysis revealed that the middle region of FAM96B is required for the limited expression of E2-2 protein. When FAM96B was expressed in ECs, the EC migration, proliferation, and tube formation were potentiated. Taken together, these findings suggest that FAM96B acts as a regulator of E2-2 through the control of its protein expression.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Proteínas Nucleares/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células COS , Proteínas Portadoras/genética , Movimiento Celular , Proliferación Celular , Cricetinae , Secuencias Hélice-Asa-Hélice , Humanos , Metaloproteínas , Ratones , Células 3T3 NIH , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Proteína 2 Similar al Factor de Transcripción 7/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA