Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Mol Med ; 28(10): e18397, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38766687

RESUMEN

Malignant insulinoma is an extremely rare type of functioning pancreatic neuroendocrine tumour with a high degree of malignancy and a high incidence of metastasis. However, it is still unclear how malignant insulinomas develop and metastasize. Serum amyloid P component (SAP), a member of the pentraxin protein family, is an acute-phase protein secreted by liver cells. The role of SAP in insulinoma and the related mechanism are still unknown. To determine the effect of SAP on insulinoma, we crossed Rip1-Tag2 mice, which spontaneously develop insulinoma, and SAP knockout (KO) mice to generate Rip1-Tag2;SAP-/- mice. We found that SAP deletion significantly promoted the growth, invasion and metastasis of malignant insulinoma through C-X-C motif chemokine ligand 12 (CXCL12) secreted by cancer-associated fibroblasts (CAFs). Further study showed that SAP deletion promoted CXCL12 secretion by CAFs through the CXCR4/p38/ERK signalling pathway. These findings reveal a novel role and mechanism of SAP in malignant insulinoma and provide direct evidence that SAP may be a therapeutic agent for this disease.


Asunto(s)
Quimiocina CXCL12 , Insulinoma , Sistema de Señalización de MAP Quinasas , Ratones Noqueados , Receptores CXCR4 , Animales , Insulinoma/metabolismo , Insulinoma/patología , Insulinoma/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Ratones , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Eliminación de Gen , Progresión de la Enfermedad , Humanos , Línea Celular Tumoral , Proliferación Celular
2.
BMC Med ; 21(1): 115, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36978108

RESUMEN

BACKGROUND: Adenoma-adenocarcinoma transition is a key feature of colorectal cancer (CRC) occurrence and is closely regulated by tumor-associated macrophages (TAMs) and CD8+ T cells. Here, we investigated the effect of the NF-κB activator 1 (Act1) downregulation of macrophages in the adenoma-adenocarcinoma transition. METHODS: This study used spontaneous adenoma-developing ApcMin/+, macrophage-specific Act1-knockdown (anti-Act1), and ApcMin/+; anti-Act1 (AA) mice. Histological analysis was performed on CRC tissues of patients and mice. CRC patients' data retrieved from the TCGA dataset were analyzed. Primary cell isolation, co-culture system, RNA-seq, and fluorescence-activated cell sorting (FACS) were used. RESULTS: By TCGA and TISIDB analysis, the downregulation of Act1 expression in tumor tissues of CRC patients negatively correlated with accumulated CD68+ macrophages in the tumor. Relative expression of EMT markers in the tumor enriched ACT1lowCD68+ macrophages of CRC patients. AA mice showed adenoma-adenocarcinoma transition, TAMs recruitment, and CD8+ T cell infiltration in the tumor. Macrophages depletion in AA mice reversed adenocarcinoma, reduced tumor amounts, and suppressed CD8+ T cell infiltration. Besides, macrophage depletion or anti-CD8a effectively inhibited metastatic nodules in the lung metastasis mouse model of anti-Act1 mice. CRC cells induced activation of IL-6/STAT3 and IFN-γ/NF-κB signaling and the expressions of CXCL9/10, IL-6, and PD-L1 in anti-Act1 macrophages. Anti-Act1 macrophages facilitated epithelial-mesenchymal-transition and CRC cells' migration via CXCL9/10-CXCR3-axis. Furthermore, anti-Act1 macrophages promoted exhaustive PD1+ Tim3+ CD8+ T cell formation. Anti-PD-L1 treatment repressed adenoma-adenocarcinoma transition in AA mice. Silencing STAT3 in anti-Act1 macrophages reduced CXCL9/10 and PD-L1 expression and correspondingly inhibited epithelial-mesenchymal-transition and CRC cells' migration. CONCLUSIONS: Act1 downregulation in macrophages activates STAT3 that promotes adenoma-adenocarcinoma transition via CXCL9/10-CXCR3-axis in CRC cells and PD-1/PD-L1-axis in CD8+ T cells.


Asunto(s)
Adenocarcinoma , Adenoma , Neoplasias Colorrectales , Animales , Ratones , Adenocarcinoma/patología , Adenoma/genética , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Terapia de Inmunosupresión , Interleucina-6 , Macrófagos/metabolismo , Macrófagos/patología , FN-kappa B/metabolismo , Humanos
3.
BMC Cancer ; 23(1): 479, 2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37237269

RESUMEN

BACKGROUND: B-cell lymphoma 2 (Bcl-2) family proteins are key regulators of apoptosis, which possess four conserved Bcl-2 homologies (BH) domains. Among the BH domains, the BH3 domain is considered as a potent 'death domain' while the BH4 domain is required for anti-apoptotic activity. Bcl-2 can be converted to a pro-apoptotic molecule through the removal or mutation of the BH4 domain. Bcl-2 is considered as an inducer of angiogenesis, which can promote tumor vascular network formation and further afford nutrients and oxygen to promote tumor progression. However, whether disrupting the function of the BH4 domain to convert Bcl-2 into a pro-apoptotic molecule could make Bcl-2 possess the potential for anti-angiogenic therapy remains to be defined. METHODS: CYD0281 was designed and synthesized according to the lead structure of BDA-366, and its function on inducing a conformational change of Bcl-2 was further evaluated via immunoprecipitation (IP) and immunofluorescence (IF) assays. Moreover, the function of CYD0281 on apoptosis of endothelial cells was analyzed via cell viability, flow cytometry, and western blotting assays. Additionally, the role of CYD0281 on angiogenesis in vitro was determined via endothelial cell migration and tube formation assays and rat aortic ring assay. Chick embryo chorioallantoic membrane (CAM) and yolk sac membrane (YSM) models, breast cancer cell xenograft tumor on CAM and in mouse models as well as the Matrigel plug angiogenesis assay were used to explore the effects of CYD0281 on angiogenesis in vivo. RESULTS: We identified a novel potent small-molecule Bcl-2-BH4 domain antagonist, CYD0281, which exhibited significant anti-angiogenic effects both in vitro and in vivo, and further inhibited breast cancer tumor growth. CYD0281 was found to induce conformational changes in Bcl-2 through the exposure of the BH3 domain and convert Bcl-2 from an anti-apoptotic molecule into a cell death inducer, thereby resulting in the apoptosis of vascular endothelial cells. CONCLUSIONS: This study has revealed CYD0281 as a novel Bcl-2-BH4 antagonist that induces conformational changes of Bcl-2 to convert to a pro-apoptotic molecule. Our findings indicate that CYD0281 plays a crucial role in anti-angiogenesis and may be further developed as a potential anti-tumor drug candidate for breast cancer. This work also provides a potential anti-angiogenic strategy for breast cancer treatment.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Embrión de Pollo , Ratones , Humanos , Ratas , Animales , Femenino , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células Endoteliales/metabolismo , Dominios Proteicos , Neoplasias de la Mama/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Apoptosis , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
4.
Exp Cell Res ; 387(1): 111756, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31811830

RESUMEN

Breast cancer (BC) is one of the most common cancers among women in both developed and developing countries with a rising incidence. Using the MMTV-PyMT transgenic mouse model and xenografted breast cancer model, we found that R5, a neutralizing antibody to Robo1, significantly inhibited BC growth and metastasis. Angiogenesis is involved in the growth and metastasis of BC. Interestingly, R5 significantly decreases microvessel density in BC tissues, and inhibits blood vessel formation and development in in vivo chick embryo chorioallantoic membrane (CAM), yolk sac membrane (YSM) and Matrigel plug models. To investigate whether its anti-breast cancer efficacy is ascribed to its direct antiangiogenic properties, xenografted breast cancer model on CAM was established. Furthermore, R5 significantly reduces the tube formation of the vascular plexus on xenografted breast tumor on CAM. R5 also suppresses the migration and the tubular structure formation of human umbilical vein endothelial cells (HUVECs) by down-regulating the expression of filamin A (FLNA). These findings show that R5 has the potential to be a promising agent for the treatment of BC by suppressing the tumor-induced angiogenesis.


Asunto(s)
Anticuerpos Neutralizantes/fisiología , Neoplasias de la Mama/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Filaminas/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Neoplasias de la Mama/metabolismo , Línea Celular , Línea Celular Tumoral , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Proteínas Roundabout
5.
Angiogenesis ; 23(3): 325-338, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32020421

RESUMEN

Breast cancer is one of the most common cancers worldwide with a rising incidence, and is the leading cause of cancer-related death among females. Angiogenesis plays an important role in breast cancer growth and metastasis. In this study, we identify decylubiquinone (DUb), a coenzyme Q10 analog, as a promising anti-breast cancer agent through suppressing tumor-induced angiogenesis. We screened a library comprising FDA-approved drugs and found that DUb significantly inhibits blood vessel formation using in vivo chick embryo chorioallantoic membrane (CAM) and yolk sac membrane (YSM) models. DUb was further identified to inhibit angiogenesis in the rat aortic ring and Matrigel plug assay. Moreover, DUb was found to suppress breast cancer growth and metastasis in the MMTV-PyMT transgenic mouse and human xenograft tumor models. To explore whether the anticancer efficacy of DUb was directly corrected with tumor-induced angiogenesis, the MDA-MB-231 breast cancer assay on the CAM was performed. Interestingly, DUb significantly inhibits the angiogenesis of breast cancer on the CAM. Brain angiogenesis inhibitor 1 (BAI1), a member of the G protein-coupled receptor (GPCR) adhesion subfamily, has an important effect on the inhibition of angiogenesis. Further studies demonstrate that DUb suppresses the formation of tubular structures by regulating the reactive oxygen species (ROS)/p53/BAI1 signaling pathway. These results uncover a novel finding that DUb has the potential to be an effective agent for the treatment of breast cancer by inhibiting tumor-induced angiogenesis.


Asunto(s)
Neoplasias de la Mama , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica , Poli(ADP-Ribosa) Polimerasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Ubiquinona/análogos & derivados , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Embrión de Pollo , Femenino , Humanos , Células MCF-7 , Metástasis de la Neoplasia , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Ubiquinona/farmacología
6.
Cancer Immunol Immunother ; 69(8): 1409-1421, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32242260

RESUMEN

MMP12 is mainly secreted by macrophages, is involved in macrophage development, and decomposes the extracellular matrix. Herein, we investigated whether macrophages would change in the intestinal tumor microenvironment after MMP12 knockout. ApcMin/+;MMP12-/-mice were obtained by crossbreeding ApcMin/+ mice with MMP12 knockout mice (MMP12-/- mice). The data showed that the number and volume of intestinal tumors were significantly increased in ApcMin/+;MMP12-/- mice compared with ApcMin/+ mice. Additionally, the tumor biomarkers CA19-9, CEA, and ß-catenin appeared relatively early in intestinal tumors in ApcMin/+;MMP12-/- mice. The results demonstrated that knocking out MMP12 accelerated the tumor growth and pathological process. On further investigation of its mechanism, the proportions of M2 macrophages in the spleen and among peritoneal macrophages were significantly up-regulated in ApcMin/+;MMP12-/- mice. Expression of M2 macrophage-related genes was up-regulated in tumor and peritoneal macrophages. The M2-related cytokine levels of IL-4 and IL-13 were increased in the serum of ApcMin/+;MMP12-/-mice. In vitro, bone marrow-derived M2 macrophages were obtained by treating bone marrow cells with IL-4 and IL-13, and these M2 macrophages secreted cytokines being changed. This finding reveals the crucial role of MMP12 in macrophage development and provides a new target for the control of macrophage polarization. Knocking out MMP12 causes intestinal M2 macrophage accumulation in tumor microenvironment, promoting the growth of intestinal tumors in ApcMin/+ mice.


Asunto(s)
Neoplasias Intestinales/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/patología , Metaloproteinasa 12 de la Matriz/fisiología , Microambiente Tumoral/inmunología , Animales , Citocinas/metabolismo , Femenino , Neoplasias Intestinales/enzimología , Neoplasias Intestinales/patología , Macrófagos/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Arterioscler Thromb Vasc Biol ; 35(2): 368-77, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25503990

RESUMEN

OBJECTIVE: An aneurysm is an inflammatory vascular condition. Phosphatidylinositol 3-kinases δ is highly expressed in leukocytes, and play a key role in innate immunity. However, the link between phosphatidylinositol 3-kinases δ and aneurysm development has not yet been elucidated. APPROACH AND RESULTS: Carotid ligation unexpectedly induced characteristic aneurysm formation beneath the ligation point in p110δ(D910A/D910A) mice (n=25; P<0.001 versus wild-type). Besides, p110δ inactivation exacerbated CaCl2-induced abdominal aortic aneurysms development. A reverse transcription polymerase chain reaction microarray revealed significant extracellular matrix components degradation and matrix metalloproteinases (MMPs) upregulation in the abdominal aorta of p110δ(D910A/D910A) mice. Similarly, the expression of both collagen I and IV was significantly decreased (n=10; P<0.05 versus wild-type) in carotid artery. Western blot assay confirmed that MMP-12 was significantly upregulated in arteries of p110δ(D910A/D910A) mice (n=10; P<0.01 versus wild-type). In vitro, p110δ inactivation marked increase peritoneal macrophages recruitment and synergistically enhance tumor necrosis factor-α-induced recruitment. A specific phosphatidylinositol 3-kinases δ inhibitor (IC87114) or genetic p110δ inactivation upregulated MMP-12 expression and c-Jun phosphorylation (n=6; P<0.05 versus wild-type macrophages). IC87114 also increased activator protein-1 DNA-binding activity (n=6; P<0.001 versus control) and enhanced the effect of tumor necrosis factor-α on activator protein-1-binding activity (n=5; P<0.01 versus tumor necrosis factor-α treatment groups). Knockdown of c-Jun suppressed the effect of the IC87114 and tumor necrosis factor-α on MMP-12 mRNA expression (n=5 in each group; P<0.01 versus scrRNA treatment groups). CONCLUSIONS: Our findings demonstrate that p110δ inactivation leads to extracellular matrix degradation in vessels and promotes aneurysm development by inducing macrophages migration and upregulating the activator protein-1/MMP-12 pathway in macrophages.


Asunto(s)
Aorta Abdominal/enzimología , Aneurisma de la Aorta Abdominal/enzimología , Traumatismos de las Arterias Carótidas/enzimología , Arteria Carótida Común/enzimología , Macrófagos Peritoneales/enzimología , Metaloproteinasa 12 de la Matriz/metabolismo , Fosfatidilinositol 3-Quinasas/deficiencia , Factor de Transcripción AP-1/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/patología , Aorta Abdominal/cirugía , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Cloruro de Calcio , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/patología , Arteria Carótida Común/cirugía , Línea Celular , Fosfatidilinositol 3-Quinasa Clase I , Modelos Animales de Enfermedad , Activación Enzimática , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica , Ligadura , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Interferencia de ARN , Transducción de Señal , Factor de Transcripción AP-1/genética , Transfección , Factor de Necrosis Tumoral alfa/farmacología
8.
Exp Cell Res ; 333(2): 261-272, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25823921

RESUMEN

Glipizide, a second-generation sulfonylurea, has been widely used for the treatment of type 2 diabetes. However, it is controversial whether or not glipizide would affect angiogenesis or vasculogenesis. In the present study, we used early chick embryo model to investigate the effect of glipizide on angiogenesis and vasculogenesis, which are the two major processes for embryonic vasculature formation as well as tumor neovascularization. We found that Glipizide suppressed both angiogenesis in yolk-sac membrane (YSM) and blood island formation during developmental vasculogenesis. Glipizide did not affect either the process of epithelial to mesenchymal transition (EMT) or mesoderm cell migration. In addition, it did not interfere with separation of smooth muscle cell progenitors from hemangioblasts. Moreover, natriuretic peptide receptor A (NPRA) has been identified as the putative target for glipizide׳s inhibitory effect on vasculogenesis. When NPRA was overexpressed or activated, blood island formation was reduced. NPRA signaling may play a crucial role in the effect of glipizide on vasculogenesis during early embryonic development.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Glipizida/farmacología , Hipoglucemiantes/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Receptores del Factor Natriurético Atrial/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Embrión de Pollo , Transición Epitelial-Mesenquimal , Gastrulación , Expresión Génica , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Saco Vitelino/irrigación sanguínea
9.
J Hepatol ; 63(6): 1413-20, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26264936

RESUMEN

BACKGROUND & AIMS: The secretory protein Slit2 and its receptor Robo1 are believed to regulate cell growth and migration. Here, we aimed to determine whether Slit2-Robo1 signaling mediates the pathogenesis of liver fibrosis. METHODS: Serum levels of Slit2 in patients with liver fibrosis were determined by ELISA. Liver fibrosis was induced in wild-type (WT), Slit2 transgenic (Slit2-Tg) and Robo1(+/-)Robo2(+/-) double heterozygotes (Robo1/2(+/-)) mice by carbon tetrachloride (CCl4). The functional contributions of Slit2-Robo1 signaling in liver fibrosis and activation of hepatic stellate cells (HSCs) were investigated using primary mouse HSCs and human HSC cell line LX-2. RESULTS: Significantly increased serum Slit2 levels and hepatic expression of Slit2 and Robo1 were observed in patients with liver fibrosis. Compared to WT mice, Slit2-Tg mice were much more vulnerable to CCl4-induced liver injury and more readily develop liver fibrosis. Development of hepatic fibrosis in Slit2-Tg mice was associated with a stronger hepatic expression of collagen I and α-smooth muscle actin (α-SMA). However, liver injury and hepatic expression of collagen I and α-SMA were attenuated in CCl4-treated Robo1/2(+/-) mice in response to CCl4 exposure. In vitro, Robo1 neutralizing antibody R5 and Robo1 siRNA downregulated phosphorylation of Smad2, Smad3, PI3K, and AKT in HSCs independent of TGF-ß1. R5 and Robo1 siRNA also inhibited the expression of α-SMA by HSCs. Finally, the protective effect of R5 on the CCl4-induced liver injury and fibrosis was further verified in mice. CONCLUSIONS: Slit2-Robo1 signaling promotes liver injury and fibrosis through activation of HSCs.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Estudios de Casos y Controles , Línea Celular , Células Cultivadas , Femenino , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Cirrosis Hepática/patología , Cirrosis Hepática Experimental/etiología , Cirrosis Hepática Experimental/metabolismo , Cirrosis Hepática Experimental/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Transducción de Señal , Proteínas Roundabout
10.
Lab Invest ; 94(7): 766-76, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24840330

RESUMEN

Slit, a neuronal guidance cue, binds to Roundabout (Robo) receptors to modulate neuronal, leukocytic, and endothelial migration. Slit has been reported to have an important effect on tumor growth and metastasis. In the current study, we evaluated the role of Slit2 in skin tumor growth and invasion in mice using a two-step chemical carcinogenesis protocol. We found that Slit2 expression correlated with the loss of basement membrane in the samples of human skin squamous cell carcinoma at different stages of disease progression. Slit2-Tg mice developed significantly more skin tumors than wild-type mice. Furthermore, the skin tumors that occurred in Slit2-Tg mice were significantly larger than those in the wild-type mice 10 weeks after 7,12-dimethylbenz[a]anthracene initiation until the end of the experiment. We also found that pathological development of the wild-type mice was delayed compared with that of Slit2-Tg mice. To further investigate the mechanism of increasing tumors in Slit2-Tg mice, we analyzed the expression of 5-bromo-2'-deoxyuridine (BrdU) in mouse skin lesions and found that the number of BrdU-positive cells and microvessel density in skin lesions were significantly higher in Slit2-Tg mice than in wild-type mice. Histological staining of PAS and type IV collagen and the colocalization of Slit2 and type IV collagen demonstrated varying degrees of loss of the basement membrane in the skin lesions from Slit2-Tg mice that were at the stage of carcinoma in situ. However, the basement membrane was well defined in the wild-type mice. In addition, MMP2, but not MMP9, was upregulated in the skin tissue of Slit2-Tg mice. Interruption of Slit2-Robo1 signaling by the antibody R5 significantly repressed the invasive capability of the squamous cell carcinoma cell line A431. Taken together, our findings reveal that Slit2 promotes DMBA/TPA-induced skin tumorigenesis by increasing cell proliferation, microvessel density, and invasive behavior of cutaneous squamous cell carcinoma, along with loss of basement membrane, by upregulation of MMP2 expression.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Cutáneas/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animales , Membrana Basal/metabolismo , Membrana Basal/patología , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Bromodesoxiuridina/metabolismo , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Proliferación Celular , Colágeno Tipo IV/metabolismo , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Invasividad Neoplásica , Proteínas del Tejido Nervioso/genética , Piel/irrigación sanguínea , Piel/metabolismo , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Acetato de Tetradecanoilforbol , Carga Tumoral
11.
Exp Mol Pathol ; 96(2): 195-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24448024

RESUMEN

Non-small cell lung cancer (NSCLC) accounts for approximately 70% of all lung cancer-related deaths worldwide. Prognostic markers are essential for the early detection of lung cancer in patients. In this study, we first identified microRNA146 (miR-146) expression in cancer cell lines using miRNA in situ hybridization (MISH) and confirmed the accuracy of MISH using q-RT-PCR. In addition, two different systems, BCIP/NBT and ELF, were used to detect the signal for a comparative analysis of the specificity of MISH. Compared to the BCIP/NBT system, the ELF detection system was more effective for MISH. Furthermore we detected the expression of miR-146 in NSCLC tissues (43 cases) and normal tissues (32 cases). Based on our results, we can conclude that miR-146 is more highly expressed in cancer tissue than normal tissue (t-test, P<0.05) and that miR-146 can predict the prognosis of NSCLC by MISH. Our findings preliminary demonstrate that MISH can be applied as a molecular diagnostic tool to determine the expression and localization of miRNAs in cancer tissues and that miR-146, determined by MISH, predicts the prognosis of NSCLC patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/biosíntesis , Pronóstico , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Humanos , Hibridación in Situ , MicroARNs/genética
12.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167114, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38447883

RESUMEN

AIMS: Exchange protein directly activated by cAMP 1 (EPAC1), a major isoform of guanine nucleotide exchange factors, is highly expressed in vascular endothelia cells and regulates angiogenesis in the retina. High intratumor microvascular densities (MVD) resulting from angiogenesis is responsible for breast cancer development. Downregulation of EPAC1 in tumor cell reduces triple-negative breast cancer (TNBC)-induced angiogenesis. However, whether Epac1 expressed in vascular endothelial cells contributes to angiogenesis and tumor development of TNBC remains elusive. MAIN METHODS: We employed NY0123, a previously identified potent EPAC inhibitor, to explore the anti-angiogenic biological role of EPAC1 in vitro and in vivo through vascular endothelial cells, rat aortic ring, Matrigel plug, and chick embryo chorioallantoic membrane (CAM) and yolk sac membrane (YSM) assays, as well as the in vivo xenograft tumor models of TNBC in both chick embryo and mice. KEY FINDINGS: Inhibiting EPAC1 in vascular endothelial cells by NY0123 significantly suppresses angiogenesis and tumor growth of TNBC. In addition, NY0123 possesses a better inhibitory efficacy than ESI-09, a reported specific EPAC inhibitor tool compound. Importantly, inhibiting EPAC1 in vascular endothelia cells regulates the typical angiogenic signaling network, which is associated with not only vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor-2 (VEGFR2) signaling, but also PI3K/AKT, MEK/ERK and Notch pathway. CONCLUSIONS: Our findings support that EPAC1 may serve as an effective anti-angiogenic therapeutic target of TNBC, and EPAC inhibitor NY0123 has the therapeutic potential to be developed for the treatment of TNBC.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido , Neovascularización Patológica , Neoplasias de la Mama Triple Negativas , Animales , Embrión de Pollo , Humanos , Ratones , Ratas , Células Endoteliales/metabolismo , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neovascularización Patológica/tratamiento farmacológico
13.
J Cell Biochem ; 114(11): 2562-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23744703

RESUMEN

Mesangial cells (MCs) proliferation and accumulation of glomerular matrix proteins such as fibronectin (FN) are the early features of diabetic nephropathy, with MCs known to upregulate matrix protein synthesis in response to high glucose. Recently, it has been found that andrographolide has renoprotective effects on diabetic nephropathy. However, the molecular mechanism underlying these effects remains unclear. Cell viability and proliferation was evaluated by MTT. FN expression was examined by immunofluorescence and immunoblotting. Activator protein-1 (AP-1) activation was assessed by immunoblotting, luciferase reporter and electrophoretic mobility shift assays. Andrographolide significantly decreased high glucose-induced cell proliferation and FN expression in MCs. Exposure of MCs to high glucose markedly stimulated the expression of phosphorylated c-jun, whereas the stimulation was inhibited by andrographolide. Plasmid pAP-1-Luc luciferase reporter assay showed that andrographolide blocked high glucose-induced AP-1 transcriptional activity. EMSA assay demonstrated that increased AP-1 binding to an AP-1 binding site at -1,029 in the FN gene promoter upon high glucose stimulation, and the binding were disrupted by andrographolide treatment. These data indicate that andrographolide suppresses high glucose-induced FN expression by inhibiting AP-1-mediated pathway.


Asunto(s)
Diterpenos/farmacología , Fibronectinas/metabolismo , Glucosa/farmacología , Células Mesangiales/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Ensayo de Cambio de Movilidad Electroforética , Fibronectinas/genética , Immunoblotting , Células Mesangiales/efectos de los fármacos , Proteínas Asociadas a Pancreatitis , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
14.
Mol Omics ; 18(2): 167-177, 2022 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-34918730

RESUMEN

A 1H NMR-based metabonomic approach was applied to monitor the alterations of serum metabolic profiles in MMTV-PyMT transgenic mice to detect the dynamic changes associated with the pathological process and explore the early-stage biomarkers. The 1H NMR spectra of sera samples from four different stages in MMTV-PyMT mice including hyperplasia, adenoma, early carcinoma and late carcinoma stages were recorded and analyzed using multivariate statistical techniques. The results showed that the increased levels of lipid and lactate, and decreased leucine/isoleucine, valine, methionine, glutamine, creatine, PC/GPC, taurine and glucose were of significance for the early carcinoma stage. As the disease progressed (late carcinoma stage), the metabolic profiles changed significantly; some were negatively regulated compared with those at the early carcinoma stage, such as lipid, leucine/isoleucine, methionine and creatine, accompanied by other new metabolite changes of alanine, pyruvate, glutamate, citrate, aspartate, myo-inositol, 3-methylhistidine and formate. It is important to note that breast cancer patients and the early carcinoma stage of MMTV-PyMT mice had some similar metabolite changes, including lipid, lactate, glutamine, creatine, taurine and glucose, which were determined to be of great value for the early clinical diagnosis of breast cancer. The findings from this study provided valuable biomarkers for the early clinical diagnosis of breast cancer, and showed the potential power of integrating NMR techniques and pattern recognition methods for the analysis of the biochemical changes under certain pathophysiological conditions.


Asunto(s)
Neoplasias de la Mama , Animales , Biomarcadores , Neoplasias de la Mama/diagnóstico , Detección Precoz del Cáncer , Femenino , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Espectroscopía de Protones por Resonancia Magnética
15.
Anticancer Agents Med Chem ; 22(9): 1735-1741, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34515012

RESUMEN

BACKGROUND: Breast cancer is one of the most common cancers worldwide among women, and angiogenesis has an important effect on its growth and metastasis. Glipizide, which is a widely used drug for type 2 diabetes mellitus, has been reported to inhibit tumor growth and metastasis by upregulating the expression of natriuretic peptide receptor A (NPRA). Atrial natriuretic peptide (ANP), the receptor of NPRA, plays an important role in angiogenesis. The purpose of this study was to explore the effect of glipizide combined with ANP on breast cancer growth and metastasis. METHODS: This study aimed at investigating the effect of glipizide combined with ANP on breast cancer. Glipizide, ANP, or glipizide combined with ANP was intraperitoneally injected into MMTV-PyMT mice. To explore whether the anticancer efficacy of glipizide combined with ANP was correlated with angiogenesis, a tube formation assay was performed. RESULTS: Glipizide combined with ANP was found to inhibit breast cancer growth and metastasis in MMTV-PyMT mice, which spontaneously develop breast cancer. Furthermore, the inhibitory effect of ANP combined with glipizide was better than that of glipizide alone. ANP combined with glipizide significantly inhibited tube formation of human umbilical vein endothelial cells (HUVECs) by suppressing vascular endothelial growth factor (VEGF)/VEGFR2 (vascular endothelial growth factor receptor 2) signaling. CONCLUSION: These results demonstrate that glipizide combined with ANP has a greater potential than glipizide alone to be repurposed as an effective agent for the treatment of breast cancer by targeting tumor-induced angiogenesis.


Asunto(s)
Neoplasias de la Mama , Diabetes Mellitus Tipo 2 , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Factor Natriurético Atrial/farmacología , Factor Natriurético Atrial/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Femenino , Glipizida/farmacología , Glipizida/uso terapéutico , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular
16.
Exp Ther Med ; 22(6): 1485, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34765026

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is outbreaking globally. SARS-CoV-2 invades host cells via angiotensin-converting enzyme II (ACE2) and causes multiple-organ injury. Autopsy studies indicated that the testis of patients with COVID-19 exhibited various degrees of spermatogenic cell reduction and injury, but the composition of ACE2-expressing cells and their proportion in the testes have remained to be determined. Recent clinical evidence suggested that the ratio of male sex hormones in males with COVID-19 was significantly changed. The present study aimed to explore whether SARS-CoV-2 is able to damage the male reproductive system. For this, the ACE2-expressing cell composition and proportion in male testes were analyzed using single-cell RNA sequencing (RNA-seq) datasets downloaded from the Gene Expression Omnibus (GEO) database and immunohistochemical (IHC) staining. The single-cell RNA-seq data indicated that ACE2 mRNA was highly expressed in myoid cells, Leydig cells and spermatogenic cells, accounting for 5.45, 1.24 and 0.423% of adult testicular cells. ACE2 mRNA-expressing Sertoli cells, spermatogenic cells and myoid cells accounted for 5.00, 0.56 and 0.73% of infant testicular cells. IHC demonstrated that ACE2 protein was also highly expressed in testicular tissues. In conclusion, the present results demonstrated that testicular injury may be missed by clinicians in patients with COVID-19 and male reproductive function should be closely followed up.

17.
Epigenomics ; 13(9): 699-713, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33876672

RESUMEN

Aim: Although studies on lncRNAs in renal fibrosis have focused on target genes and functions of lncRNAs, a comprehensive interaction analysis of lncRNAs is lacking. Materials & methods: Differentially expressed genes in renal fibrosis were screened, and the interaction between lncRNAs and miRNAs was searched. Results: We constructed a ceRNA network associated with renal fibrosis, by which we found the transcription factor Creb5, a target gene of lncRNA Gas5 that might regulate extracellular Fn1 deposition. Conclusion: Our study not only provides a theoretical basis for the ceRNA regulation mechanism of Gas5 but also provides experimental evidence supporting the use of Gas5 targeting in the treatment of renal fibrosis.


Asunto(s)
Proteína de Unión al Elemento de Respuesta al AMP Cíclico/genética , Fibronectinas/metabolismo , Riñón/patología , ARN Largo no Codificante/genética , Animales , Línea Celular , Fibrosis , Masculino , Ratones Endogámicos C57BL , Regulación hacia Arriba
18.
Front Cell Dev Biol ; 9: 643525, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34249905

RESUMEN

Tumor growth and metastasis are responsible for breast cancer-related mortality. Andrographolide (Andro) is a traditional anti-inflammatory drug used in the clinic that inhibits NF-κB activation. Recently, Andro has been found in the treatment of various cancers. Andro inhibits breast cell proliferation and invasion and induces apoptosis via activating various signaling pathways. Therefore, the underlying mechanisms with regard to the antitumor effects of Andro still need to be further confirmed. Herein, a MMTV-PyMT spontaneous luminal-like breast cancer lung metastatic transgenic tumor model was employed to estimate the antitumor effects of Andro on breast cancer in vivo. Andro significantly inhibited tumor growth and metastasis in MMTV-PyMT mice and suppressed the cell proliferation, migration, and invasion of MCF-7 breast cancer cells in vitro. Meanwhile, Andro significantly inhibited the expression of NF-κB, and the downregulated NF-κB reduced miR-21-5p expression. In addition, miR-21-5p dramatically inhibited the target gene expression of programmed cell death protein 4 (PDCD4). In the current study, we demonstrated the potential anticancer effects of Andro on luminal-like breast cancer and indicated that Andro inhibits the expression of miR-21-5p and further promotes PDCD4 via NF-κB suppression. Therefore, Andro could be an antitumor agent for the treatment of luminal-like breast cancer in the clinic.

19.
Front Pharmacol ; 12: 804265, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35177983

RESUMEN

Colorectal cancer (CRC) is one of the leading causes of cancer-related death worldwide. Decylubiquinone (DUb), a coenzyme Q10 analog, was reported to inhibit breast cancer growth and metastasis by us. However, the influence of DUb on CRC remains unclear. Herein, we found that DUb significantly inhibited CRC growth in the patient-derived xenograft (PDX) and CT26 xenograft models. DUb was further identified to significantly suppress CRC cell proliferation, colony formation, migration and invasion in a dose-dependent manner, while not inhibiting CRC cell apoptosis from flow cytometry assay. Sirtuin2 (SIRT2), a member of the sirtuin protein family, plays a critical role in growth and metastasis in various cancers. Moreover, DUb inhibited CRC progression by upregulating SIRT2. These findings reveal that DUb has the potential to a novel drug for the treatment of CRC by inhibiting CRC cell proliferation.

20.
Biomaterials ; 266: 120432, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33069116

RESUMEN

Gastrointestinal (GI) cancers are among the most lethal malignancies. The treatment of advanced-stage GI cancer involves standard chemotherapeutic drugs, such as docetaxel, as well as targeted therapeutics and immunomodulatory agents, all of which are only moderately effective. We here show that Π electron-stabilized polymeric micelles based on PEG-b-p(HPMAm-Bz) can be loaded highly efficiently with docetaxel (loading capacity up to 23 wt%) and potentiate chemotherapy responses in multiple advanced-stage GI cancer mouse models. Complete cures and full tumor regression were achieved upon intravenously administering micellar docetaxel in subcutaneous gastric cancer cell line-derived xenografts (CDX), as well as in CDX models with intraperitoneal and lung metastases. Nanoformulated docetaxel also outperformed conventional docetaxel in a patient-derived xenograft (PDX) model, doubling the extent of tumor growth inhibition. Furthermore, micellar docetaxel modulated the tumor immune microenvironment in CDX and PDX tumors, increasing the ratio between M1-and M2-like macrophages, and toxicologically, it was found to be very well-tolerated. These findings demonstrate that Π electron-stabilized polymeric micelles loaded with docetaxel hold significant potential for the treatment of advanced-stage GI cancers.


Asunto(s)
Antineoplásicos , Neoplasias Gastrointestinales , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Docetaxel , Portadores de Fármacos , Electrones , Neoplasias Gastrointestinales/tratamiento farmacológico , Ratones , Micelas , Polietilenglicoles , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA