Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Intervalo de año de publicación
1.
Stem Cell Res Ther ; 15(1): 98, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38581019

RESUMEN

BACKGROUND: In vitro chondrogenesis of mesenchymal stromal cells (MSCs) driven by the essential chondro-inducer transforming growth factor (TGF)-ß is instable and yields undesired hypertrophic cartilage predisposed to bone formation in vivo. TGF-ß can non-canonically activate bone morphogenetic protein-associated ALK1/2/3 receptors. These have been accused of driving hypertrophic MSC misdifferentiation, but data remained conflicting. We here tested the antihypertrophic capacity of two highly specific ALK1/2/3 inhibitors - compound A (CompA) and LDN-212854 (LDN21) - in order to reveal potential prohypertrophic contributions of these BMP/non-canonical TGF-ß receptors during MSC in vitro chondrogenesis. METHODS: Standard chondrogenic pellet cultures of human bone marrow-derived MSCs were treated with TGF-ß and CompA (500 nM) or LDN21 (500 nM). Daily 6-hour pulses of parathyroid hormone-related peptide (PTHrP[1-34], 2.5 nM, from day 7) served as potent antihypertrophic control treatment. Day 28 samples were subcutaneously implanted into immunodeficient mice. RESULTS: All groups underwent strong chondrogenesis, but GAG/DNA deposition and ACAN expression were slightly but significantly reduced by ALK inhibition compared to solvent controls along with a mild decrease of the hypertrophy markers IHH-, SPP1-mRNA, and Alkaline phosphatase (ALP) activity. When corrected for the degree of chondrogenesis (COL2A1 expression), only pulsed PTHrP but not ALK1/2/3 inhibition qualified as antihypertrophic treatment. In vivo, all subcutaneous cartilaginous implants mineralized within 8 weeks, but PTHrP pretreated samples formed less bone and attracted significantly less haematopoietic marrow than ALK1/2/3 inhibitor groups. CONCLUSIONS: Overall, our data show that BMP-ALK1/2/3 inhibition cannot program mesenchymal stromal cells toward stable chondrogenesis. BMP-ALK1/2/3 signalling is no driver of hypertrophic MSC misdifferentiation and BMP receptor induction is not an adverse prohypertrophic side effect of TGF-ß that leads to endochondral MSC misdifferentiation. Instead, the prohypertrophic network comprises misregulated PTHrP/hedgehog signalling and WNT activity, and a potential contribution of TGF-ß-ALK4/5-mediated SMAD1/5/9 signalling should be further investigated to decide about its postulated prohypertrophic activity. This will help to successfully engineer cartilage replacement tissues from MSCs in vitro and translate these into clinical cartilage regenerative therapies.


Asunto(s)
Células Madre Mesenquimatosas , Proteína Relacionada con la Hormona Paratiroidea , Animales , Humanos , Ratones , Células Cultivadas , Condrocitos/metabolismo , Condrogénesis , Proteínas Hedgehog/genética , Hipertrofia/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Factor de Crecimiento Transformador beta/metabolismo
2.
Front Immunol ; 15: 1302489, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38476229

RESUMEN

Background: Pre-neutrophils, while developing in the bone marrow, transcribe the Inhba gene and synthesize Activin-A protein, which they store and release at the earliest stage of their activation in the periphery. However, the role of neutrophil-derived Activin-A is not completely understood. Methods: To address this issue, we developed a neutrophil-specific Activin-A-deficient animal model (S100a8-Cre/Inhba fl/fl mice) and analyzed the immune response to Influenza A virus (IAV) infection. More specifically, evaluation of body weight and lung mechanics, molecular and cellular analyses of bronchoalveolar lavage fluids, flow cytometry and cell sorting of lung cells, as well as histopathological analysis of lung tissues, were performed in PBS-treated and IAV-infected transgenic animals. Results: We found that neutrophil-specific Activin-A deficiency led to exacerbated pulmonary inflammation and widespread hemorrhagic histopathology in the lungs of IAV-infected animals that was associated with an exuberant production of neutrophil extracellular traps (NETs). Moreover, deletion of the Activin-A receptor ALK4/ACVR1B in neutrophils exacerbated IAV-induced pathology as well, suggesting that neutrophils themselves are potential targets of Activin-A-mediated signaling. The pro-NETotic tendency of Activin-A-deficient neutrophils was further verified in the context of thioglycollate-induced peritonitis, a model characterized by robust peritoneal neutrophilia. Of importance, transcriptome analysis of Activin-A-deficient neutrophils revealed alterations consistent with a predisposition for NET release. Conclusion: Collectively, our data demonstrate that Activin-A, secreted by neutrophils upon their activation in the periphery, acts as a feedback mechanism to moderate their pro-NETotic tendency and limit the collateral tissue damage caused by neutrophil excess activation during the inflammatory response.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Neumonía , Animales , Ratones , Humanos , Neutrófilos , Pulmón/patología , Neumonía/metabolismo , Gripe Humana/patología , Activinas/metabolismo
3.
Chem Biodivers ; 20(2): e202200817, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36604308

RESUMEN

A compound library of sixty six linear compounds, eleven representatives of six molecular families: (E)- and (Z)-isomers of alk-4-en-1-ols, alk-4-enals, and methyl alk-4-enoates, was prepared by combinatorial syntheses to allow the creation of a mass spectral database directly usable for their identification in GC/MS analyses. We demonstrate here that compound libraries can be prepared by combinatorial syntheses using long linear synthetic sequences, i. e., eight step in the case of 4-enals. The resulting mixtures of homologues are still perfectly exploitable to deliver the requested information such as clean mass spectra and good gas chromatographic retention indices.


Asunto(s)
Bibliotecas de Moléculas Pequeñas , Humanos , Cromatografía de Gases y Espectrometría de Masas/métodos , Cromatografía de Gases , Espectrometría de Masas , Isomerismo
4.
Biomed Pharmacother ; 154: 113573, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35987161

RESUMEN

Although many research have found that colchicine has general therapeutic effect in cardiovascular disease, the therapeutic mechanism in atrial fibrillation has not been clearly studied. To explore whether colchicine plays a role in the treatment of AF by reducing myocardial fibrosis, we performed a series of studies. Rat models of AF were induced by Ach-CaCl2 to assess the therapeutic effect of colchicine at doses of 0.8 mg/kg on the duration of AF rhythm, degree of myocardial fibrosis, and secretion of inflammatory factors in the serum. RNA-Seq was also performed to elucidate the possible mechanisms by which colchicine might reduce the alleviation of myocardial fibrosis associated with AF. These studies showed that colchicine reduced the duration of AF and the degree of fibrosis in the left atrium and that it significantly reduced the secretion of TGFß1, activin A, collagen I, and collagen III. These results suggest that colchicine may reduce myocardial fibrosis by (1) inhibiting the TGFß1/ALK5 and activin A/ALK4 fibrosis pathways; (2) inhibiting the activation, phenotypic transformation, and apoptosis resistance of myocardial fibroblasts; and (3) reducing the synthesis of inflammatory factors and collagen.


Asunto(s)
Fibrilación Atrial , Cardiomiopatías , Animales , Fibrilación Atrial/metabolismo , Cardiomiopatías/metabolismo , Colchicina/metabolismo , Colchicina/farmacología , Colchicina/uso terapéutico , Colágeno Tipo I/metabolismo , Fibrosis , Atrios Cardíacos/patología , Ratas , Ratas Sprague-Dawley
5.
Dev Dyn ; 251(1): 226-234, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34816532

RESUMEN

BACKGROUND: Precise gastrulation is essential for formation of functional bodies in cnidarians and bilaterians. Previously, by using an alk4/5/7-specific inhibitor, we showed that transforming growth factor-beta (TGF-ß)-alk4/5/7 signaling pathway is important for correct gut bending in sea urchin embryos. However, it is still unclear where functional TGF-ß signals are received in embryos for correct gut bending because details of the spatiotemporal expression pattern of alk4/5/7 have not been reported. RESULTS: We revealed that alk4/5/7 are expressed from the 2-cell to early pluteus stage throughout the entire body, including the invaginating gut. To investigate whether TGF-ß signals directly received in endoderm are required for correct gut bending, we made chimeras in which alk4/5/7 translation was inhibited only in endomesoderm lineage. As a result, the gut of the chimeric embryos did not bend precisely, in contrast to the control chimeras. CONCLUSION: We conclude that direct TGF-ß signaling to the endoderm via alk4/5/7 pathway regulates correct gut bending. However, TGF-ß-alk4/5/7 pathway is not related to mouth opening because the mouth is formed without TGF-ß signaling to the endoderm. This research contributes to understanding the mechanisms leading to the proper positioning of the end of the archenteron for forming a through-gut, which is commonly needed for bilaterians.


Asunto(s)
Gastrulación , Erizos de Mar , Animales , Endodermo , Gástrula , Gastrulación/fisiología , Transducción de Señal/fisiología
6.
Aging (Albany NY) ; 13(13): 17473-17488, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34214050

RESUMEN

BACKGROUND: Inflammation process is an important determinant for subsequent changes in cardiac function and remodeling after acute myocardial infarction (MI). Recent studies have implicated that ALK4 haplodeficiency improves cardiac function after MI. However, it remains unknown if the beneficial effects are partly attributed to ALK4 haplodeficiency-induced modulation on inflammatory response in the inflammatory phase of MI. In this research, we aimed to explore the mechanism of ALK4 haplodeficiency in the inflammatory stage of MI. METHODS: ALK4, CD16, and CD14 were detected in peripheral blood mononuclear cells (PBMCs) isolated from MI patients and healthy volunteers. ALK4 haplodeficiency (ALK4+/-) mice and wild-type (WT) littermates were randomly divided into the sham group and the MI group. Inflammation cytokines and chemokines were measured. Echocardiography and intracardiac electrophysiological recordings were performed on the 3rd day and the 7th day after MI operation. ALK4 expression and inflammation cytokines were also detected in LPS- or IL-4-stimulated bone marrow-derived macrophages (BMDM) from the ALK4+/- mice and WT littermates. RESULTS: ALK4 gene expression in circulating monocytes of MI patients was higher than that in those of healthy volunteers. Cardiac inflammation and vulnerability of ventricular arrhythmia after acute myocardial injury are significantly alleviated in ALK4+/- mice as compared to WT littermates. On the 3rd day post-MI, the level of M1 macrophages were decreased in ALK4+/- mice as compared to WT littermates, while the level of M2 macrophages were increased on the 7th day post-MI. BMDM isolated from ALK4+/- mice displayed reduced secretion of pro-inflammation cytokines after stimulation by LPS in hypoxic condition and increased secretion of anti-inflammation cytokines after stimulation by IL-4. As a result, the haplodeficiency of ALK4 might be responsible for reduced inflammation response in the post-MI stage. CONCLUSIONS: ALK4 haplodeficiency reduces cardiac inflammation, improves cardiac function, and finally reduces the vulnerability of ventricular arrhythmia in the inflammatory stage after MI.


Asunto(s)
Receptores de Activinas Tipo I/genética , Arritmias Cardíacas/etiología , Arritmias Cardíacas/genética , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Miocarditis/patología , Animales , Estimulación Cardíaca Artificial , Citocinas/metabolismo , Ecocardiografía , Proteínas Ligadas a GPI/genética , Voluntarios Sanos , Humanos , Receptores de Lipopolisacáridos/genética , Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de IgG/genética
7.
Immunity ; 54(2): 308-323.e6, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33421362

RESUMEN

Th17 cells are known to exert pathogenic and non-pathogenic functions. Although the cytokine transforming growth factor ß1 (TGF-ß1) is instrumental for Th17 cell differentiation, it is dispensable for generation of pathogenic Th17 cells. Here, we examined the T cell-intrinsic role of Activin-A, a TGF-ß superfamily member closely related to TGF-ß1, in pathogenic Th17 cell differentiation. Activin-A expression was increased in individuals with relapsing-remitting multiple sclerosis and in mice with experimental autoimmune encephalomyelitis. Stimulation with interleukin-6 and Activin-A induced a molecular program that mirrored that of pathogenic Th17 cells and was inhibited by blocking Activin-A signaling. Genetic disruption of Activin-A and its receptor ALK4 in T cells impaired pathogenic Th17 cell differentiation in vitro and in vivo. Mechanistically, extracellular-signal-regulated kinase (ERK) phosphorylation, which was essential for pathogenic Th17 cell differentiation, was suppressed by TGF-ß1-ALK5 but not Activin-A-ALK4 signaling. Thus, Activin-A drives pathogenic Th17 cell differentiation, implicating the Activin-A-ALK4-ERK axis as a therapeutic target for Th17 cell-related diseases.


Asunto(s)
Activinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Inflamación Neurogénica/inmunología , Células Th17/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Activinas/genética , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Transducción de Señal
8.
Front Cell Dev Biol ; 9: 765007, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34977017

RESUMEN

The epicardium, the mesothelial layer covering the heart, is a crucial cell source for cardiac development and repair. It provides cells and biochemical signals to the heart to facilitate vascularization and myocardial growth. An essential element of epicardial behavior is epicardial epithelial to mesenchymal transition (epiMT), which is the initial step for epicardial cells to become motile and invade the myocardium. To identify targets to optimize epicardium-driven repair of the heart, it is vital to understand which pathways are involved in the regulation of epiMT. Therefore, we established a cell culture model for human primary adult and fetal epiMT, which allows for parallel testing of inhibitors and stimulants of specific pathways. Using this approach, we reveal Activin A and ALK4 signaling as novel regulators of epiMT, independent of the commonly accepted EMT inducer TGFß. Importantly, Activin A was able to induce epicardial invasion in cultured embryonic mouse hearts. Our results identify Activin A/ALK4 signaling as a modulator of epicardial plasticity which may be exploitable in cardiac regenerative medicine.

9.
BMC Urol ; 20(1): 49, 2020 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366240

RESUMEN

BACKGROUND: Inhibition of angiogenesis in prostatic cancer could be a brand-new method to suppress tumour progression. Nodal/ALK4 has been associated with vascularization in many cancers. However, the relationship between and role of Nodal/ALK4 and miR-185 in human prostatic cancer is still unknown. METHODS: Prostatic cancer DU145 cells and LNCaP cells were used to investigate the angiogenic effect induced by Nodal and the anti-angiogenic roles of miR-185. Colony formation assay, MTT assay, transwell assay and tube formation assay were used to explore cell proliferation, migration and tube-forming ability, respectively. A luciferase reporter assay confirmed the binding relationship between miR-185 and ALK4. The expression levels of miR-185, ALK4 and VEGF were detected by qRT-PCR and Western blotting. The effects of miR-185 and Nodal in prostate cancer were also investigated in animal experiments. RESULTS: VEGF expression was increased in DU145 cells and LNCaP cells after Nodal incubation, and Nodal activated the proliferation ability of prostatic cancer cells and the migration and tube-forming ability of human umbilical vein endothelial cells (HUVECs), which were all inhibited by treatment with the Nodal inhibitor SB431524. Bioinformatics analysis and luciferase assay were used to verify miR-185 as a target of ALK4. Prostatic cancer cell proliferation was inhibited by overexpression of miR-185, which was shown to regulate the migration and angiogenesis of HUVECs by targeting ALK4 for suppression. miR-185 also showed a significant inverse correlation with Nodal treatment and reversed the angiogenic effects induced by Nodal. More importantly, for the first time, xenograft experiments indicated that overexpression of miR-185 suppressed tumour development. CONCLUSION: The Nodal/ALK4 pathway is important in the angiogenesis of prostate cancer and can be inhibited by targeting miR-185 to downregulate ALK4. These findings provide a new perspective on the mechanism of prostate cancer formation.


Asunto(s)
Receptores de Activinas Tipo I/fisiología , MicroARNs/fisiología , Proteína Nodal/fisiología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/genética , Humanos , Masculino , Neovascularización Patológica , Neoplasias de la Próstata/patología , Células Tumorales Cultivadas
10.
Biochem J ; 477(8): 1391-1407, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32215602

RESUMEN

Bicyclic peptides assembled around small organic scaffolds are gaining an increasing interest as new potent, stable and highly selective therapeutics because of their uncommon ability to specifically recognize protein targets, of their small size that favor tissue penetration and of the versatility and easiness of the synthesis. We have here rationally designed bicyclic peptides assembled around a common tri-bromo-methylbenzene moiety in order to mimic the structure of the CFC domain of the oncogene Cripto-1 and, more specifically, to orient in the most fruitful way the hot spot residues H120 and W123. Through the CFC domain, Cripto-1 binds the ALK4 receptor and other protein partners supporting uncontrolled cell growth and proliferation. Soluble variants of CFC have the potential to inhibit these interactions suppressing the protein activity. A CFC analog named B3 binds ALK4 in vitro with an affinity in the nanomolar range. Structural analyses in solution via NMR and CD show that B3 has rather flexible conformations, like the parent CFC domain. The functional effects of B3 on the Cripto-1-positive NTERA cancer cell line have been evaluated showing that both CFC and B3 are cytotoxic for the cells and block the Cripto-1 intracellular signaling. Altogether, the data suggest that the administration of the soluble CFC and of the structurally related analog has the potential to inhibit tumor growth.


Asunto(s)
Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Péptidos/química , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Secuencias de Aminoácidos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Diseño de Fármacos , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Espectroscopía de Resonancia Magnética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Péptidos/farmacología
11.
Mol Cell Endocrinol ; 505: 110731, 2020 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-31982478

RESUMEN

In the mammalian ovary, the proteolysis of the extracellular matrix is dynamically regulated by plasminogen activator and plasminogen activator inhibitor (PAI), and it is a critical event that influences various physiological and pathological processes. Activin A is a member of the transforming growth factor-ß superfamily and is expressed at a high level in human luteal cells that play an essential role in the regulation of the luteal function. At present, it is not known whether activin A can regulate the expression and production of PAI in human granulosa lutein (hGL) cells. The present study aimed to examine the effects of activin A on the expression and production of intraovarian PAI-1 and the underlying molecular mechanisms. Using primary and immortalized hGL cells as the cell model, we demonstrated that activin A upregulated the expression of PAI-1 and increased the production of PAI-1 in an autocrine/paracrine manner. Additionally, using a dual inhibition approach (molecular inhibitors and siRNA-mediated knockdown), we showed that this biological function is mediated by the ALK4-mediated SMAD3-SMAD4-dependent signaling pathway. Our findings suggest that activin A may be involved in the regulation of luteal function via the induction of PAI-1 expression and an increase in PAI-1 production.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Activinas/metabolismo , Células Lúteas/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteínas Smad/metabolismo , Regulación hacia Arriba , Benzamidas/farmacología , Línea Celular , Células Cultivadas , Dioxoles/farmacología , Femenino , Humanos , Modelos Biológicos , Fosforilación/efectos de los fármacos , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/efectos de los fármacos
12.
Mol Cell Endocrinol ; 493: 110485, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31185247

RESUMEN

As one of the members of the transforming growth factor-ß (TGF-ß) superfamily, activin A plays an important role in regulating follicular development and oocyte maturation. Pentraxin 3 (PTX3) is the key component that promotes the process of cumulus expansion during mammalian ovulation. At present, the regulation of PTX3 expression in human granulosa cells remains largely unknown. This study aimed to examine the effects of activin A on the expression of PTX3 in human granulosa-lutein (hGL) cells and to investigate the underlying molecular mechanisms. Using an established immortalized hGL cell line (SVOG) and primary hGL cells as study models, we demonstrated that activin A significantly increased the phosphorylation of SMAD2 and SMAD3, which suppressed the expression of PTX3 at both the mRNA and protein levels. Additionally, these effects induced by activin A were completely reversed by pretreatment with the TGF-ß type I receptor inhibitor SB431542 and knockdown of ALK4. Furthermore, knockdown of SMAD2, SMAD3, or SMAD4 completely reversed the activin A-induced suppressive effects on PTX3 expression. Notably, the ChIP analyses demonstrated that phosphorylated SMADs could bind to human PTX3 promoter. Collectively, our results showed that the ALK4-SMAD2/3-SMAD4 signaling pathway most likely mediates the suppressive effect of activin A on PTX3 expression in hGL cells.


Asunto(s)
Activinas/farmacología , Proteína C-Reactiva/genética , Proteína C-Reactiva/metabolismo , Células Lúteas/citología , Componente Amiloide P Sérico/genética , Componente Amiloide P Sérico/metabolismo , Receptores de Activinas Tipo I/genética , Benzamidas/farmacología , Células Cultivadas , Dioxoles/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Lúteas/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Proteína Smad4/metabolismo
13.
Onco Targets Ther ; 11: 6589-6597, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30323631

RESUMEN

BACKGROUND: Bladder cancer is the most common malignant tumor of the urinary tract. We aimed to explore the biological role and molecular mechanism of Nodal in bladder cancer. MATERIALS AND METHODS: The expression of Nodal in bladder cancer tissues and cells was determined by quantitative real-time polymerase chain reaction. The effect of silencing of Nodal on cell proliferation, clone formation, and migration and invasion was evaluated by MTT cell proliferation assay, colony formation, and transwell assays, respectively. Western blot analysis was employed to detect the expression of proliferation- and invasion-related proteins and proteins involved in ALK/Smad signaling. RESULTS: We found that the expression of Nodal was significantly increased in bladder cancer tissues and cell lines. Downregulation of Nodal effectively weakened cell proliferation, clone formation, and cell migration and invasion abilities. The protein expression levels of CDC6, E-cadherin, MMP-2, and MMP-9 were also altered by downregulation of Nodal. Knockdown of Nodal also blocked the expression of ALK4, ALK7, Smad2, and Smad4, which are involved in ALK/Smad signaling. Additionally, the ALK4/7 receptor blocker SB431542 reversed the promotive effects of Nodal overexpression on bladder cancer cell proliferation, migration, and invasion. CONCLUSION: Our study indicated that Nodal functions as an oncogene by regulating cell proliferation, migration, and invasion in bladder cancer via the ALK/Smad signaling pathway, thereby providing novel insights into its role in bladder cancer treatment.

14.
J Am Heart Assoc ; 7(16): e008842, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30369314

RESUMEN

Background Activin receptor-like kinase 4 ( ALK 4) is highly expressed in mammal heart. Atrial fibrillation ( AF ) is closely related to ventricular pressure overload. Because pressure overload increases atrial pressure and leads to atrial remodeling, it would be informative to know whether ALK 4 exerts potential effects on atrial remodeling and AF vulnerability in a pressure-overload model. Methods and Results Wild-type littermates and ALK 4+/- mice were subjected to abdominal aortic constriction or a sham operation. After 4 or 8 weeks, echocardiographic and hemodynamic measurements were performed, and inducibility of AF was tested. The hearts were divided into atria and ventricles and then were fixed in formalin for staining, or they were weighted and snap-frozen for quantitative real-time polymerase chain reaction and Western blot analysis. Compared with wild-type littermates, ALK 4+/- mice demonstrated a similar extent of atrial hypertrophy but significantly suppressed atrial fibrosis at 8 weeks post-abdominal aortic constriction. ALK 4 haplodeficiency partially blocked abdominal aortic constriction-induced upregulation of monocyte chemotactic protein 1 and interleukin-6, and the increased chemotaxin of macrophages. ALK 4 haplodeficiency also blunted a reduction of connexin 40 and redistribution of connexin 43 from the intercalated disk to the lateral membranes, thereby improving localized conduction abnormalities. Meanwhile, ALK 4 haplodeficiency inhibited abdominal aortic constriction-induced decreased INa, ICa-L and IK1 densities as well as the accompanying action potential duration shortening. Mechanistically, ALK 4 haploinsufficiency resulted in the suppression of Smad2/3 activity in this model. Conclusions Our results demonstrate that ALK 4 haplodeficiency ameliorates atrial remodeling and vulnerability to AF in a pressure-overload model through inactivation of the Smad2/3 pathway, suggesting that ALK 4 might be a potential therapeutic target in combating pressure overload-induced AF .


Asunto(s)
Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Fibrilación Atrial/genética , Remodelación Atrial/genética , Cardiomegalia , Miocitos Cardíacos/metabolismo , Anciano , Animales , Aorta Abdominal/cirugía , Quimiocina CCL2/metabolismo , Factores Quimiotácticos/metabolismo , Conexina 43/metabolismo , Conexinas/metabolismo , Femenino , Fibrosis , Predisposición Genética a la Enfermedad , Haploinsuficiencia , Sistema de Conducción Cardíaco , Humanos , Hipertensión , Interleucina-6/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Técnicas de Placa-Clamp , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Proteína alfa-5 de Unión Comunicante
15.
Biomed Pharmacother ; 98: 440-445, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29278854

RESUMEN

Aristaless-like homeobox 4 (ALK4) is a member of ALK proteins family and plays an important role in tumorigenesis. However, the expression and function of ALK4 in glioma remain largely unknown. The aim of our study was to elucidate its expression pattern in human glioma tissues and cell lines, as well as its functions in glioma cells. Our results demonstrated that ALK4 was lowly expressed in human glioma tissues and cell lines. Additionally, overexpression of ALK4 significantly suppressed the proliferation, migration and invasion of glioma cells, as well as inhibited the epithelial-mesenchymal transition (EMT) phenotype in glioma cells. Furthermore, overexpression of ALK4 significantly downregulated the phosphorylation levels of JAK2 and STAT3 in U87 cells. STAT3 inhibitor (Niclosamide) obviously enhanced ALK4-inhibted glioma cell proliferation and invasion. In conclusion, we demonstrated that overexpression of ALK4 suppressed glioma cell proliferation, migration and invasion through the inactivation of JAK/STAT3 signaling pathway. Thus, ALK4 may be a potential therapeutic target for the treatment of glioma.


Asunto(s)
Receptores de Activinas Tipo I/genética , Movimiento Celular/genética , Proliferación Celular/genética , Glioma/genética , Janus Quinasa 2/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/genética , Carcinogénesis/genética , Línea Celular Tumoral , Regulación hacia Abajo/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Invasividad Neoplásica/genética
16.
Basic Res Cardiol ; 112(4): 47, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28639003

RESUMEN

Atrial fibrosis, the hallmark of structural remodeling associated with atrial fibrillation (AF), is characterized by abnormal proliferation of atrial fibroblasts and excessive deposition of extracellular matrix. Transforming growth factor-ß1 (TGF-ß1)/activin receptor-like kinase 5 (ALK5)/Smad2/3/4 pathway has been reported to be involved in the process. Recent studies have implicated both activin A and its specific downstream component activin receptor-like kinase 4 (ALK4) in stimulating fibrosis in non-cardiac organs. We recently reported that ALK4 haplodeficiency attenuated the pressure overload- and myocardial infarction-induced ventricular fibrosis. However, the role of activin A/ALK4 in the pathogenesis of atrial fibrosis and vulnerability to AF remains unknown. Our study provided experimental and clinical evidence for the involvement of activin A and ALK4 in the pathophysiology of atrial fibrosis and AF. Patients with AF had higher activin A and ALK4 expression in atriums as compared to individuals devoid of AF. After angiotensin-II (Ang-II) stimulation which mimicked atrial fibrosis progression, ALK4-deficient mice showed lower expression of ALK4 in atriums, reduced activation of atrial fibroblasts, blunted atrial enlargement and atrial fibrosis, and further reduced AF vulnerability upon right atrial electrophysiological studies as compared to wild-type littermates. Moreover, we found that apart from the well-known TGF-ß1/ALK5 pathway, the activation of activin A/ALK4/smad2/3 pathway played an important role in the pathogenesis of Ang-II-mediated atrial fibrosis and inducibility of AF, suggesting that targeting ALK4 might be a potential therapy for atrial fibrosis and AF.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Activinas/metabolismo , Fibrilación Atrial/metabolismo , Atrios Cardíacos/patología , Adulto , Anciano , Angiotensina II/toxicidad , Animales , Fibrilación Atrial/patología , Femenino , Fibrosis , Atrios Cardíacos/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad
17.
Arch Biochem Biophys ; 608: 34-41, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27592306

RESUMEN

Nodal is a member of transforming growth factor beta (TGF-ß) superfamily. Nodal promotes the self-renewal of human cancer stem cells (CSCs) and triggers carcinogenesis of human cancers via an autocrine manner through Smad2/3 pathway. In our study, generation of Nodal-overexpressed cancer cells was constructed, and the effect of Nodal on the stem cell marker Oct-4 was evaluated by overexpression or blocked Nodal/ALKs signaling pathway in non-small cell lung cancer cells A549 and prostate cancer cells PC3. Functionally, Nodal also increased the proliferation via the ß-catenin nuclear translocation. This increase was attributed to GSK-3ß dephosphorylating, and activin receptor-like kinase 4/7 (ALK4/7) played a major role in human cancer cells. Our study provides a positive understanding of Nodal function in cancer cells and suggests a potential novel target for clinical therapeutic research.


Asunto(s)
Transporte Activo de Núcleo Celular , Regulación Neoplásica de la Expresión Génica , Proteína Nodal/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Neoplasias de la Próstata/metabolismo , beta Catenina/metabolismo , Células A549 , Receptores de Activinas Tipo I/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Citoplasma/metabolismo , Humanos , Masculino , Transducción de Señal , Transfección
18.
Eur J Pharmacol ; 789: 319-327, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27477354

RESUMEN

Activin A is a key regulator of cardiac fibrosis. However, little is known about the mechanisms by which it contributes to cardiac fibrosis. Our study explored the effects of activin A on proliferation and differentiation of adult rat cardiac fibroblasts (CFs) via the activin A receptor, activin receptor-like kinase 4 (ALK4). CF proliferation was measured by CCK8 and EdU assays, while differentiation, fibrosis and signaling were measured by western blot analysis of α-smooth muscle actin, collagen type I, phosphorylated extracellular signal-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (p38-MAPK) expression. Activin A levels were measured by ELISA and western blot analysis. We demonstrated that CFs express activin A and its expression was significantly enhanced by angiotensin II (Ang II), but follistatin (activin A inhibitor) significantly reversed Ang II-induced activin A upregulation, CF proliferation, differentiation, collagen type I expression as well as ERK1/2 and p38-MAPK pathways activation. Conversely, recombinant activin A largely increased these parameters in both the presence and absence of Ang II. Interestingly, p38-MAPK (SB203580) and ALK4 (SB431542) inhibitors significantly reduced all activin A-mediated responses; however, an ERK1/2 inhibitor (PD98059) could only significantly reduce CF proliferation and collagen type I expression but not differentiation. Importantly, the most significant effects were observed in the presence vs. absence of Ang II. Thus, activin A promotes basal and Ang II-induced CF proliferation and differentiation via ALK4, and the effects are partly mediated through the ERK1/2 and p38-MAPK pathways. These data suggest that activin A is a potential therapeutic target for cardiac fibrosis.


Asunto(s)
Activinas/farmacología , Diferenciación Celular/efectos de los fármacos , Fibroblastos/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Receptores de Activinas Tipo I/metabolismo , Angiotensina II/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Miocardio/citología , Ratas , Ratas Sprague-Dawley
19.
Artículo en Inglés | MEDLINE | ID: mdl-22645523

RESUMEN

Nodal is a member of the transforming growth factor-ß (TGF-ß) superfamily that plays critical roles during embryogenesis. Recent studies in ovarian, breast, prostate, and skin cancer cells suggest that Nodal also regulates cell proliferation, apoptosis, and invasion in cancer cells. However, it appears to exert both tumor-suppressing and tumor-promoting effects, depending on the cell type. To further understand the role of Nodal in tumorigenesis, we examined the effect of Nodal in glioblastoma cell growth and spheroid formation using U87 cell line. Treatment of U87 with recombinant Nodal significantly increased U87 cell growth. In U87 cells stably transfected with the plasmid encoding Nodal, Smad2 phosphorylation was strongly induced and cell growth was significantly enhanced. Overexpression of Nodal also resulted in tight spheroid formation. On the other hand, the cells stably transfected with Nodal siRNA formed loose spheroids. Nodal is known to signal through activin receptor-like kinase 4 (ALK4) and ALK7 and the Smad2/3 pathway. To determine which receptor and Smad mediate the growth promoting effect of Nodal, we transfected siRNAs targeting ALK4, ALK7, Smad2, or Smad3 into Nodal-overexpressing cells and observed that cell growth was significantly inhibited by ALK4, ALK7, and Smad3 siRNAs. Taken together, these findings suggest that Nodal may have tumor-promoting effects on glioblastoma cells and these effects are mediated by ALK4, ALK7, and Smad3.

20.
Immune Netw ; 11(4): 196-202, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22039367

RESUMEN

BACKGROUND: B cell-activating factor belonging to the TNF family (BAFF) is primarily expressed by macrophages and dendritic cells, and stimulates B cell proliferation, differentiation, survival, and Ig production. In the present study, we explored the effect of activin A on BAFF expression by APCs. METHODS: To investigate the effect of activin A on BAFF expression by mouse APCs, we measured the level of BAFF expression at the transcriptional and protein levels using RT-PCR and ELISA. RESULTS: Activin A markedly enhanced BAFF expression in mouse macrophages and dendritic cells at both the transcriptional and protein levels. SB431542, an activin receptor-like kinase 4 (ALK4) inhibitor, completely abrogated activin A-induced BAFF transcription. Furthermore, overexpression of DN-Smad3 abolished activin-induced BAFF expression at the transcriptional and protein levels. CONCLUSION: These results demonstrate that activin A can enhance BAFF expression through ALK4-Smad3 pathway.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA