Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 11(17)2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36078170

RESUMEN

The TGF-ß/Smad3 signaling pathway is an important process in the pathogenesis of kidney fibrosis. However, the molecular mechanisms are not completely elucidated. The current study examined the functional role of S100A4 in regulating TGF-ß/Smad3 signaling in fibroblast activation and kidney fibrosis development. S100A4 was upregulated in the kidney in a murine model of renal fibrosis induced by folic acid nephropathy. Further, S100A4 was predominant in the tubulointerstitial cells of the kidney. Pharmacological inhibition of S100A4 with niclosamide significantly attenuated fibroblast activation, decreased collagen content, and reduced extracellular matrix protein expression in folic acid nephropathy. Overexpression of S100A4 in cultured renal fibroblasts significantly facilitated TGF-ß1-induced activation of fibroblasts by increasing the expression of α-SMA, collagen-1 and fibronectin. In contrast, S100A4 knockdown prevented TGF-ß1-induced activation of fibroblast and transcriptional activity of Smad3. Mechanistically, S100A4 interacts with Smad3 to stabilize the Smad3/Smad4 complex and promotes their translocation to the nucleus. In conclusion, S100A4 facilitates TGF-ß signaling via interaction with Smad3 and promotes kidney fibrosis development. Manipulating S100A4 may provide a beneficial therapeutic strategy for chronic kidney disease.


Asunto(s)
Fibroblastos , Insuficiencia Renal Crónica , Proteína de Unión al Calcio S100A4 , Factor de Crecimiento Transformador beta1 , Animales , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibrosis , Ácido Fólico/metabolismo , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología , Ratones , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/patología , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo
2.
Toxicol Appl Pharmacol ; 449: 116134, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35724704

RESUMEN

Colorectal cancer (CRC) is the world's second most common cause of cancer-related death. Novel treatments are still urgently needed. S100 calcium-binding protein A4 (S100A4) was demonstrated to be an anticancer therapeutic target. Herein, we found that higher S100A4 expression was associated with a poorer prognosis in publicly available cohorts and a Taiwanese CRC patient cohort. To identify repurposed S100A4 inhibitors, we mined the Connectivity Map (CMap) database for clinical drugs mimicking the S100A4-knockdown gene signature. Ingenol mebutate, derived from the sap of the plant Euphorbia peplus, is approved as a topical treatment for actinic keratosis. The CMap analysis predicted ingenol mebutate as a potent S100A4 inhibitor. Indeed, both messenger RNA and protein levels of S100A4 were attenuated by ingenol mebutate in human CRC cells. In addition, CRC cells with higher S100A4 expressions and/or the wild-type p53 gene were more sensitive to ingenol mebutate, and their migration and invasion were inhibited by ingenol mebutate. Therefore, our results suggest the repurposing of ingenol mebutate for treating CRC by targeting S100A4.


Asunto(s)
Neoplasias Colorrectales , Diterpenos , Proteína de Unión al Calcio S100A4 , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Diterpenos/farmacología , Diterpenos/uso terapéutico , Reposicionamiento de Medicamentos , Humanos , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Proteína de Unión al Calcio S100A4/genética
3.
Cardiovasc Res ; 118(1): 141-155, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33135065

RESUMEN

AIMS: During atherosclerosis, smooth muscle cells (SMCs) accumulate in the intima where they switch from a contractile to a synthetic phenotype. From porcine coronary artery, we isolated spindle-shaped (S) SMCs exhibiting features of the contractile phenotype and rhomboid (R) SMCs typical of the synthetic phenotype. S100A4 was identified as a marker of R-SMCs in vitro and intimal SMCs, in pig and man. S100A4 exhibits intra- and extracellular functions. In this study, we investigated the role of extracellular S100A4 in SMC phenotypic transition. METHODS AND RESULTS: S-SMCs were treated with oligomeric recombinant S100A4 (oS100A4), which induced nuclear factor (NF)-κB activation. Treatment of S-SMCs with oS100A4 in combination with platelet-derived growth factor (PDGF)-BB induced a complete SMC transition towards a pro-inflammatory R-phenotype associated with NF-κB activation, through toll-like receptor-4. RNA sequencing of cells treated with oS100A4/PDGF-BB revealed a strong up-regulation of pro-inflammatory genes and enrichment of transcription factor binding sites essential for SMC phenotypic transition. In a mouse model of established atherosclerosis, neutralization of extracellular S100A4 decreased area of atherosclerotic lesions, necrotic core, and CD68 expression and increased α-smooth muscle actin and smooth muscle myosin heavy chain expression. CONCLUSION: We suggest that the neutralization of extracellular S100A4 promotes the stabilization of atherosclerotic plaques. Extracellular S100A4 could be a new target to influence the evolution of atherosclerotic plaques.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Enfermedades de la Aorta/tratamiento farmacológico , Aterosclerosis/tratamiento farmacológico , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Placa Aterosclerótica , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Actinas/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Becaplermina/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Cadenas Pesadas de Miosina/metabolismo , Fenotipo , Proteína de Unión al Calcio S100A4/metabolismo , Proteína de Unión al Calcio S100A4/farmacología , Transducción de Señal , Miosinas del Músculo Liso/metabolismo , Sus scrofa , Receptor Toll-Like 4/metabolismo
4.
J Cell Biol ; 220(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34328510

RESUMEN

Ferroptosis is a form of iron-dependent regulated cell death driven by uncontrolled lipid peroxidation. Mitochondria are double-membrane organelles that have essential roles in energy production, cellular metabolism, and cell death regulation. However, their role in ferroptosis has been unclear and somewhat controversial. In this Perspective, I summarize the diverse metabolic processes in mitochondria that actively drive ferroptosis, discuss recently discovered mitochondria-localized defense systems that detoxify mitochondrial lipid peroxides and protect against ferroptosis, present new evidence for the roles of mitochondria in regulating ferroptosis, and outline outstanding questions on this fascinating topic for future investigations. An in-depth understanding of mitochondria functions in ferroptosis will have important implications for both fundamental cell biology and disease treatment.


Asunto(s)
Ferroptosis/genética , GTP Ciclohidrolasa/antagonistas & inhibidores , Hierro/metabolismo , Mitocondrias/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Sistema de Transporte de Aminoácidos y+/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Células Cultivadas , Ferroptosis/efectos de los fármacos , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Regulación de la Expresión Génica , Glutatión/antagonistas & inhibidores , Glutatión/biosíntesis , Humanos , Peroxidación de Lípido/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Oxidantes/farmacología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Proteína de Unión al Calcio S100A4/genética , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal , Ubiquinona/antagonistas & inhibidores , Ubiquinona/metabolismo
5.
J Neuroinflammation ; 18(1): 132, 2021 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-34118929

RESUMEN

BACKGROUND: An increasing number of studies evidences that amyotrophic lateral sclerosis (ALS) is characterized by extensive alterations in different cell types and in different regions besides the CNS. We previously reported the upregulation in ALS models of a gene called fibroblast-specific protein-1 or S100A4, recognized as a pro-inflammatory and profibrotic factor. Since inflammation and fibrosis are often mutual-sustaining events that contribute to establish a hostile environment for organ functions, the comprehension of the elements responsible for these interconnected pathways is crucial to disclose novel aspects involved in ALS pathology. METHODS: Here, we employed fibroblasts derived from ALS patients harboring the C9orf72 hexanucleotide repeat expansion and ALS patients with no mutations in known ALS-associated genes and we downregulated S100A4 using siRNA or the S100A4 transcriptional inhibitor niclosamide. Mice overexpressing human FUS were adopted to assess the effects of niclosamide in vivo on ALS pathology. RESULTS: We demonstrated that S100A4 underlies impaired autophagy and a profibrotic phenotype, which characterize ALS fibroblasts. Indeed, its inhibition reduces inflammatory, autophagic, and profibrotic pathways in ALS fibroblasts, and interferes with different markers known as pathogenic in the disease, such as mTOR, SQSTM1/p62, STAT3, α-SMA, and NF-κB. Importantly, niclosamide in vivo treatment of ALS-FUS mice reduces the expression of S100A4, α-SMA, and PDGFRß in the spinal cord, as well as gliosis in central and peripheral nervous tissues, together with axonal impairment and displays beneficial effects on muscle atrophy, by promoting muscle regeneration and reducing fibrosis. CONCLUSION: Our findings show that S100A4 has a role in ALS-related mechanisms, and that drugs such as niclosamide which are able to target inflammatory and fibrotic pathways could represent promising pharmacological tools for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Niclosamida/farmacología , Niclosamida/uso terapéutico , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis/tratamiento farmacológico , Fibrosis/prevención & control , Humanos , Inflamación/tratamiento farmacológico , Inflamación/prevención & control , Ratones , Mutación , FN-kappa B/metabolismo , Proteína FUS de Unión a ARN/genética , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
6.
Int J Oncol ; 58(3): 397-408, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33650647

RESUMEN

A synthetic peptide that blocks the interaction between the metastasis­enhancing calcium­binding protein, S100A4, and its effector protein, methionine aminopeptidase 2 (MetAP2) (the NBD peptide), was previously demonstrated to inhibit the angiogenesis of endothelial cells, leading to the regression of human prostate cancer in a xenograft model. However, the effects of the NBD peptide on the malignant properties of cancer cells that express S100A4 remain to be elucidated. The present study demonstrates that the NBD peptide inhibits the invasiveness and metastasis of highly metastatic human mammary carcinoma cells. The introduction of the peptide into MDA­MB­231 variant cells resulted in the suppression of matrix degradation in a gelatin invadopodia assay and invasiveness in a Matrigel invasion assay. In line with these results, the peptide significantly downregulated the expression of matrix metalloproteinase (MMP)­14 (MT1­MMP). Mechanistic analysis of the downregulation of MMP­14 revealed the suppression of the expression of the transcription factor, specificity protein 1 (Sp1), but not that of nuclear factor (NF)­κB, early growth response 1 (EGR1) or ELK3, all of which were reported to be involved in transcriptional regulation of the MMP­14 gene. At the same time, evidence suggested that the NBD peptide also suppressed Sp1 and MMP­14 expression levels in MDA­MB­468 cells. Importantly, the intravenous administration of the NBD peptide encapsulated in liposomes inhibited pulmonary metastasis from mammary gland tumors in mice with xenograft tumors. These results indicate that the NBD peptide can suppress malignant tumor growth through the suppression of the Sp1/MMP­14 axis. Taken together, these results reveal that the NBD peptide acts on not only endothelial cells, but also on tumor cells in an integrated manner, suggesting that the peptide may prove to be a promising cancer therapeutic peptide drug.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Péptidos/farmacología , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Administración Intravenosa , Animales , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Metaloproteinasa 14 de la Matriz/metabolismo , Metionil Aminopeptidasas/genética , Ratones , Péptidos/genética , Péptidos/uso terapéutico , Dominios y Motivos de Interacción de Proteínas/genética , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer Ther ; 19(12): 2598-2611, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32999046

RESUMEN

S100A4 oncoprotein plays a critical role during prostate cancer progression and induces immunosuppression in host tissues. We hypothesized that S100A4-regulated oncogenic activity in immunosuppressed prostate tumors promotes growth of neoplastic cells, which are likely to become aggressive. In the current study, we investigated whether biopsy-S100A4 gene alteration independently predicts the outcome of disease in patients and circulatory-S100A4 is druggable target for treating immunosuppressive prostate cancer. Aided by DECIPHER-genomic test, we show biopsy-S100A4 overexpression as predictive of (i) poor ADT response and (ii) high risk of mortality in 228 radical prostatectomy-treated patients. Furthermore, analysis of tumor genome data of more than 1,000 patients with prostate cancer (PRAD/SU2C/FHCRC studies) validated the association of S100A4-alteration to poor survival and metastasis. We show that increased serum-S100A4 levels are associated to the prostate cancer progression in patients. The prerequisite for metastasis is the escape of tumor cells via vascular system. We show that extracellular-S100A4 protein as a growth factor induces vascular transmigration of prostate cancer cells and bone demineralization thus forms an ideal target for therapies for treating prostate cancer. By employing surface plasmon resonance and isothermal titration calorimetry, we show that mab6B12 antibody interacts with and neutralizes S100A4 protein. When tested for therapeutic efficacy, the mab6B12 therapy reduced the (i) osteoblastic demineralization of bone-derived MSCs, (ii) S100A4-target (NFκB/MMP9/VEGF) levels in prostate cancer cells, and (iii) tumor growth in a TRAMPC2 syngeneic mouse model. The immuno-profile analysis showed that mAb6B12-therapy (i) shifted Th1/Th2 balance (increased Stat4+/T-bet+ and decreased GATA2+/CD68+/CD45+/CD206+ cells); (ii) modulated cytokine levels in CD4+ T cells; and (iii) decreased levels of IL5/6/12/13, sTNFR1, and serum-RANTES. We suggest that S100A4-antibody therapy has clinical applicability in treating immunosuppressive prostate cancer in patients.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Inmunomodulación/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Antineoplásicos Inmunológicos/farmacología , Biomarcadores de Tumor , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Humanos , Biopsia Líquida , Recuento de Linfocitos , Masculino , Pronóstico , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/etiología , Proteína de Unión al Calcio S100A4/sangre , Proteína de Unión al Calcio S100A4/genética , Resultado del Tratamiento
8.
Gene ; 749: 144679, 2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32330536

RESUMEN

BACKGROUND: Hepatocellular carcinoma is one of the most common cancers worldwide. HBV-related HCC has characteristics of faster progression and worse prognosis. Previous studies have confirmed that HBx protein plays numbers of important roles in development of HBV-HCC. However, the molecular mechanism of carcinogenicity of HBx is still not well documented. METHODS: Firstly, a HCC cell line over-expressing HBx was established and its function was verified. Subsequently, the differentially expressed genes were detected by transcriptome sequencing technology and use the Western Blot technology to detect the up-regulated genes in HBx overexpressed cells, and the functional correlation of the genes was analyzed. Finally, tissue microarray was used to correlate up-regulated gene with clinical follow-up data to verify correlation with clinical prognosis. RESULTS: Over-expression of HBx could promote cell proliferation, and over-expression of HBx could up-regulate the expression of S100A4 protein. ShRNA experiments showed that HBx promoted cell proliferation by upregulating the expression of S100A4. IFN-α2b can down-regulate the expression of S100A4 and inhibit the proliferation of HCC cells. The expression of S100A4 in cancer was significantly up-regulated compared with adjacent tissues, and was also significantly associated with tumors volume, the expression of PD-L1 and the survival time of patients with HCC. CONCLUSION: In general, S100A4 may be an effective therapeutic target for HBV-HCC. And the connection between S100A4 and HBV are not clear yet. This study may play a guiding role in the future clinical treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteína de Unión al Calcio S100A4/metabolismo , Transactivadores/metabolismo , Animales , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Proliferación Celular , Estimación de Kaplan-Meier , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Regulación hacia Arriba , Proteínas Reguladoras y Accesorias Virales
9.
Cells ; 8(10)2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31623154

RESUMEN

S100A4, belonging to a large multifunctional S100 protein family, is a Ca2+-binding protein with a significant role in stimulating the motility of cancer and immune cells, as well as in promoting pro-inflammatory properties in different cell types. In the CNS, there is limited information concerning S100A4 presence and function. In this study, we analyzed the expression of S100A4 and the effect of the S100A4 transcriptional inhibitor niclosamide in murine activated primary microglia. We found that S100A4 was strongly up-regulated in reactive microglia and that niclosamide prevented NADPH oxidase 2, mTOR (mammalian target of rapamycin), and NF-κB (nuclear factor-kappa B) increase, cytoskeletal rearrangements, migration, and phagocytosis. Furthermore, we found that S100A4 was significantly up-regulated in astrocytes and microglia in the spinal cord of a transgenic rat SOD1-G93A model of amyotrophic lateral sclerosis. Finally, we demonstrated the increased expression of S100A4 also in fibroblasts derived from amyotrophic lateral sclerosis (ALS) patients carrying SOD1 pathogenic variants. These results ascribe S100A4 as a marker of microglial reactivity, suggesting the contribution of S100A4-regulated pathways to neuroinflammation, and identify niclosamide as a possible drug in the control and attenuation of reactive phenotypes of microglia, thus opening the way to further investigation for a new application in neurodegenerative conditions.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/metabolismo , Microglía/citología , Microglía/efectos de los fármacos , Niclosamida/uso terapéutico , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Adulto , Esclerosis Amiotrófica Lateral/inmunología , Animales , Western Blotting , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Masculino , Ratones , Microglía/inmunología , Microglía/metabolismo , Microscopía Fluorescente , Persona de Mediana Edad , FN-kappa B/metabolismo , Fagocitosis/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína de Unión al Calcio S100A4/metabolismo , Superóxido Dismutasa-1 , Serina-Treonina Quinasas TOR/metabolismo
10.
Biochem Biophys Res Commun ; 516(4): 1123-1129, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31284952

RESUMEN

Methionine aminopeptidase 2 (MetAP2) is one of the effector proteins of S100A4, a metastasis-associated calcium-binding protein. This interaction is involved in angiogenesis. The region of MetAP2 that interacts with S100A4 includes amino acids 170 to 208. A peptide corresponding to this region, named as NBD, has potent anti-angiogenic activity and suppresses tumor growth in a xenograft cancer model. However, the binding mode of NBD to S100A4 was totally unknown. Here we describe our analysis of the relationship between the inhibitory activity and the structure of NBD, which adopts a characteristic helix-turn-helix structure as shown by X-ray crystallographic analysis, and peptide fragments of NBD. We conducted physicochemical analyses of the interaction between S100A4 and the peptides, including surface plasmon resonance, microscale thermophoresis, and circular dichroism, and performed docking/molecular dynamics simulations. Active peptides had stable secondary structures, whereas inactive peptides had a little secondary structure. A computational analysis of the interaction mechanism led to the design of a peptide smaller than NBD, NBD-ΔN10, that possessed inhibitory activity. Our study provides a strategy for design for a specific peptide inhibitor against S100A4 that can be applied to the discovery of inhibitors of other protein-protein interactions.


Asunto(s)
Metionil Aminopeptidasas/química , Péptidos/química , Péptidos/farmacología , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Metionil Aminopeptidasas/metabolismo , Simulación del Acoplamiento Molecular , Unión Proteica , Estructura Secundaria de Proteína , Proteína de Unión al Calcio S100A4/metabolismo
11.
J Med Virol ; 91(12): 2083-2092, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31359441

RESUMEN

Viral myocarditis (VMC) is an inflammatory cardiac disease caused by coxsackievirus B3 (CVB3) that leads to heart failure or sudden death. However, efficient therapeutic strategies for VMC remain lacking. Ginkgo biloba extract was previously demonstrated to have anti-inflammatory activity and had been used in prevention and therapy of some cardiovascular diseases (ie myocardial infarction), indicating Ginkgo biloba extract may be a potential drug for the treatment of VMC. This study was, for the first time, to investigate the intervention effects of Ginkgo biloba extract on VMC model mice and explore its potential mechanisms. As a result, VMC mice model was successfully established by CVB3 infection, exhibiting significantly higher viral titer, serum creatine kinase isoenzyme level, heart weight/body weight ratio, histopathologic scores, collagen volume fraction (CVF), and significantly increased expression of S100A4 and matrix metalloproteinase-3 (MMP-3) at protein and messenger RNA levels compared with the control group. Also, the expression of S100A4 and MMP-3/CVF was positively correlated. Ginkgo biloba extract treatment significantly reversed the trend in all the above parameters. Thus, Ginkgo biloba extract may be a promising therapeutic approach against VMC because it improved myocardial injury and alleviated the degree of myocardial fibrosis through suppression of S100A4 and MMP-3.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enterovirus Humano B/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Miocarditis/tratamiento farmacológico , Extractos Vegetales/uso terapéutico , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Animales , Infecciones por Coxsackievirus/tratamiento farmacológico , Ginkgo biloba , Masculino , Metaloproteinasa 3 de la Matriz , Ratones , Ratones Endogámicos BALB C , Miocarditis/virología
12.
Cell Physiol Biochem ; 49(3): 1143-1162, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30196299

RESUMEN

BACKGROUND/AIMS: Anaplastic thyroid cancer (ATC), with 25% BRAFV600E mutation, is one of the most lethal human malignancies that currently has no effective therapy. Vemurafenib, a BRAFV600E inhibitor, has shown promise in clinical trials, including ATC patients, but is being hampered by the acquisition of drug resistance. Therefore, combination therapy that includes BRAFV600E inhibition and avoids resistance is a clinical need. METHODS: ATC cell lines 8505C (BRAFV600E/mt), SW1736 (BRAFV600E/mt), KAT18 (BRAFV600E/wt) and Cal-62(BRAFV600E/wt) cells were used in the study. The ability of S100A knockout or /and in combination with the BRAF inhibitor vemurafenib on growth, apoptosis, invasion and apoptosis in ATC cells in vitro was demonstrated by MTT and BrdUrd incorporation assay, Annexin-V-FITC staining analyzed by flow cytometry, Transwell migration and Matrigel invasion assay. S100A4,pERK1/2, pAKT and pROCK1/2 protein was detected by western blot assay; Small molecule inhibitors of Y27632, U0126, MK-2206 and constitutively active forms of pCDNA-Myc-pERK, pCMV6-HA-Akt, pCMV-RhoA were employed, and the mechanistic studies were performed. We assessed the efficiency of in vivo combination treatment with S100A4 knockout and Vemurafenib on tumors. RESULTS: S100A4 knockout induced apoptosis and reduced proliferation by inactivation of pAKT and pERK signals, and inhibited invasion and migration by inactivation of pAKT and RhoA/ROCK1/2 signals in 8505C or Cal-62 cells in vitro, and vice versa in SW1736 and KAT18 cells. Vemurafenib did not affect apoptosis of both 8505C and SW1736 cells, but reduced proliferation via arresting cell cycle, and promoted cell migration and invasion in vitro. Combination treatment with S100A4 knockdown and vemurafenib reduced cell proliferation, migration and invasion in vitro compared to the S100A4 knockdown or Vemurafenib alone. Vemurafenib treatment resulted in a transient inhibition of pERK expression and gradually activation of pAKT expression, but quickly recovery from ERK1/2 activation inhibition by vemurafenib treatment in 4 h for SW1736 and 8505C cells. Combined treatment completely inhibited ERK1/2 and AKT activation during 48 h. In an in vivo mouse model of SW1736 and 8505C, vemurafenib treatment alone did not significantly inhibit tumor growth in both of the tumors, but inhibited tumor growth in combined groups. CONCLUSION: Our results show S100A4 knockout alone inhibits ATC cells (rich endogenous S100A4) survival and invasion, regardless of the BRAFV600E status, and potentiates the effect of vemurafenib on tumor regression in vitro and in vivo. In addition, S100A4 knockout potently inhibits the recovery from ERK1/2 activation inhibition and the AKT activation following vemurafenib treatment and reversed the vemurafenib resistance. This therapeutic combination may be of benefit in patients with ATC.


Asunto(s)
Antineoplásicos/uso terapéutico , Indoles/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Proteína de Unión al Calcio S100A4/metabolismo , Sulfonamidas/uso terapéutico , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Indoles/farmacología , Ratones , Ratones Noqueados , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Proteína de Unión al Calcio S100A4/genética , Sulfonamidas/farmacología , Carcinoma Anaplásico de Tiroides/metabolismo , Carcinoma Anaplásico de Tiroides/patología , Vemurafenib
13.
Ann Plast Surg ; 81(2): 163-169, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29846215

RESUMEN

Recent researches have indicated that S100A4 participates in tissue fibrosis, whereas calcimycin inhibits this process as a novel S100A4 transcription inhibitor. However, the relationship and mechanisms between calcimycin and S100A4 in keloid fibroblasts (KFs) remain unknown. The present research was aimed to evaluate the effect of calcimycin on S100A4 expression and pathogenesis in KFs. Keloid fibroblasts were cultured and exposed to different concentrations of calcimycin in the absence or presence of transforming growth factor ß1 (TGF-ß1). The results showed that the expression of S100A4 was significantly increased in keloid derived fibroblasts compared with normal skin fibroblasts. Calcimycin depressed S100A4 in a concentration- and time-dependent manner. Moreover, calcimycin suppressed TGF-ß1-induced collagen type I, fibronectin, and α-smooth muscle actin expression and cell viability in cultured KFs. Furthermore, calcimycin modulated expression of TGF-ß/Smad target genes Smad7 and phosphorylation of TGF-ß1-induced Smad2/3. This research for the first time confirmed the presence of S100A4 in KFs. Calcimycin inhibits the expression of S100A4, as well as KF proliferation and migration and extracellular matrix (ECM) synthesis. Taken together, these results indicate that calcimycin might be a therapeutic candidate to keloid or other related fibrotic disorders.


Asunto(s)
Calcimicina/farmacología , Ionóforos de Calcio/farmacología , Fibroblastos/efectos de los fármacos , Queloide/metabolismo , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo , Adolescente , Adulto , Anciano , Biomarcadores/metabolismo , Western Blotting , Calcimicina/uso terapéutico , Ionóforos de Calcio/uso terapéutico , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas In Vitro , Queloide/tratamiento farmacológico , Queloide/patología , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
14.
Cell Commun Signal ; 16(1): 2, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29329589

RESUMEN

BACKGROUND: Fusion of breast cancer cells with tumor-associated populations of the microenvironment including mesenchymal stroma/stem-like cells (MSC) represents a rare event in cell communication whereby the metastatic capacity of those hybrid cells remains unclear. METHODS: Functional changes were investigated in vitro and in vivo following spontaneous fusion and hybrid cell formation between primary human MSC and human MDA-MB-231 breast cancer cells. Thus, lentiviral eGFP-labeled MSC and breast cancer cells labeled with mcherry resulted in dual-fluorescing hybrid cells after co-culture. RESULTS: Double FACS sorting and single cell cloning revealed two different aneuploid male hybrid populations (MDA-hyb1 and MDA-hyb2) with different STR profiles, pronounced telomerase activities, and enhanced proliferative capacities as compared to the parental cells. Microarray-based mRNA profiling demonstrated marked regulation of genes involved in epithelial-mesenchymal transition and increased expression of metastasis-associated genes including S100A4. In vivo studies following subcutaneous injection of the breast cancer and the two hybrid populations substantiated the in vitro findings by a significantly elevated tumor growth of the hybrid cells. Moreover, both hybrid populations developed various distant organ metastases in a much shorter period of time than the parental breast cancer cells. CONCLUSION: Together, these data demonstrate spontaneous development of new tumor cell populations exhibiting different parental properties after close interaction and subsequent fusion of MSC with breast cancer cells. This formation of tumor hybrids contributes to continuously increasing tumor heterogeneity and elevated metastatic capacities.


Asunto(s)
Transformación Celular Neoplásica , Células Madre Mesenquimatosas/metabolismo , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Proteína de Unión al Calcio S100A4/genética , Proteína de Unión al Calcio S100A4/metabolismo , Telomerasa/metabolismo
15.
Cell Biol Int ; 41(10): 1103-1109, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28675500

RESUMEN

FAM107B expression was decreased in stomach cancer and many other kinds of cancer. The forced expression of FAM107B in HeLa cells diminished proliferation in response to growth factors, suggesting that FAM107B might play important roles in many types of cancers. But the mechanisms underlying the decreased expression of FAM107B in cancers are not clear, the functional significance needs to be further clarified. Our previous findings from cDNA microarray showed that there are 179 differentially expressed genes after S100A4 inhibition in gastric cancer cells MGC803. FAM107B was an upregulated one among them. In the present study, we confirmed that FAM107B expression was upregulated in MGC803 cells after S100A4 inhibition by qRT-PCR. We demonstrated for the first time that FAM107B was downregulated by S100A4. The results from CCK-8 and transwell assay showed that FAM107B inhibition by siRNA led to significantly increased proliferation and migrating abilities of MGC803 cells, respectively, indicating that FAM107B plays important roles in inhibiting the proliferation and migration of MGC803 cells. The rescue experiment showed that FAM107B-siRNA transfection reversed the reduced proliferation and migration abilities induced by S100A4 inhibition in the cells. These findings suggest that, as a downstream effector, FAM107B at least partly mediates the effect of S100A4 on the proliferation and migration of MGC803 cells. In conclusion, we first provide experimental evidence suggesting that FAM107B was downregulated by S100A4 in gastric cancer MGC803 cells. And FAM107B at least partially mediates the biological effect of S100A4 in the cells.


Asunto(s)
Proteínas Nucleares/biosíntesis , Proteína de Unión al Calcio S100A4/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Apoptosis/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Regulación hacia Abajo , Genes Supresores de Tumor , Células HeLa/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Transfección
16.
Int J Oncol ; 49(2): 559-68, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27278086

RESUMEN

Many studies have revealed that S100A4 is involved in cancer progression by affecting a variety of biological functions. Our previous study showed that S100A4 influences many biological properties of gastric cancer cells; however, the underlying mechanisms are far from clear. In this study, we used cDNA microarray analysis to investigate the global alterations in gene expression in MGC803 gastric cancer cells after siRNA-mediated S100A4 inhibition. Among the total genes investigated, 179 differentially expressed genes (38 upregulated and 141 downregulated) were detected in S100A4-siRNA transfected MGC803 cells compared with NC-siRNA transfected cells. We focused on the GDF15 gene, which was significantly downregulated after S100A4 inhibition. ChIP studies showed that the S100A4 protein binds to the GDF15 promoter, implicating S100A4 in GDF15 regulation at the transcriptional level. GDF15 overexpression promoted CSC-like properties of MGC803 cells, such as spheroid and soft-agar colony forming abilities. S100A4 inhibition suppressed the CSC-like properties of the cells, whereas, GV141-GDF15 vector transfection reversed these effects. Our results suggest that S100A4 influences the CSC-like properties of MGC803 gastric cancer cells by regulating GDF15 expression.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/biosíntesis , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteína de Unión al Calcio S100A4/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Regiones Promotoras Genéticas , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Proteína de Unión al Calcio S100A4/genética , Transfección
17.
Oncotarget ; 7(23): 34630-42, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27127879

RESUMEN

S100A4 (metastasin-1), a metastasis-associated protein and marker of the epithelial to mesenchymal transition, contributes to several hallmarks of cancer and has been implicated in the progression of several types of cancer. However, the impacts of S100A4 signaling in lung cancer progression and its potential use as a target for therapy in lung cancer have not been properly explored. Using established lung cancer cell lines, we demonstrate that S100A4 knockdown reduces cell proliferation, invasion and three-dimensional invasive growth, while overexpression of S100A4 increases invasive potential. In patient-derived tissues, S100A4 is preferentially elevated in lung adenocarcinoma. This elevation is associated with lymphovascular invasion and decreased overall survival. In addition, depletion of S100A4 by shRNA inhibits NF-κB activity and decreases TNFα-induced MMP9 expression. Furthermore, inhibition of the NF-κB/MMP9 axis decreases lung carcinoma invasive potential. Niclosamide, a reported inhibitor of S100A4, blocks expression and function of S100A4 with a reduction in proliferation, invasion and NF-κB-mediated MMP9 expression. Collectively, this study highlights the importance of the S100A4/NF-κB/MMP9 axis in lung cancer invasion and provides a rationale for targeting S100A4 to combat lung cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Niclosamida/farmacología , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Proteína de Unión al Calcio S100A4/metabolismo , Células A549 , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , Neoplasias Pulmonares/patología , Metaloproteinasa 9 de la Matriz/biosíntesis , FN-kappa B/metabolismo , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Pronóstico , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteína de Unión al Calcio S100A4/genética , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...