Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 249
Filtrar
1.
Molecules ; 27(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35056817

RESUMEN

Melatonin is a hormone secreted by the pineal gland that is involved in the biorhythm of reproductive activities. The present study investigated the inhibitory effects of melatonin on osteoclastogenesis in RAW 264.7 cells according to changes in V-ATPase and the corresponding inhibition of the MAPK and NFATc1 signaling processes. METHODS: the cytotoxic effect of melatonin was investigated by MTT assay. Osteoclast differentiation and gene expression of osteoclast-related factors were confirmed via TRAP staining, pit formation assay, immunofluorescence imaging, western blot, and real-time PCR. RESULTS: melatonin was found to inactivate the p38 and JNK of MAP kinase in RAW264.7 cells treated with RANKL and treated with a combination RANKL and melatonin for 1, 3, and 5 days. The melatonin treatment group showed a reduction in osteoclastogenesis transcription factors and ATP6v0d2 gene expression. CONCLUSIONS: melatonin inhibits osteoclast differentiation and cell fusion by inhibiting the expression of Atp6v0d2 through the inactivation of MAPK and NFATc1 signaling in RANKL-stimulated RAW264.7 macrophages. The findings of the present study suggest that melatonin could be a suitable therapy for bone loss and imply a potential role of melatonin in bone health.


Asunto(s)
Melatonina/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Factores de Transcripción NFATC/antagonistas & inhibidores , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Osteoclastos/citología , Osteogénesis , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores , Animales , Antioxidantes/farmacología , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular , Células Cultivadas , Regulación hacia Abajo , Regulación de la Expresión Génica , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , FN-kappa B/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Células RAW 264.7
2.
Immunity ; 55(2): 224-236.e5, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34995475

RESUMEN

During gram-negative septicemia, interactions between platelets and neutrophils initiate a detrimental feedback loop that sustains neutrophil extracellular trap (NET) induction, disseminated intravascular coagulation, and inflammation. Understanding intracellular pathways that control platelet-neutrophil interactions is essential for identifying new therapeutic targets. Here, we found that thrombin signaling induced activation of the transcription factor NFAT in platelets. Using genetic and pharmacologic approaches, as well as iNFATuation, a newly developed mouse model in which NFAT activation can be abrogated in a cell-specific manner, we demonstrated that NFAT inhibition in activated murine and human platelets enhanced their activation and aggregation, as well as their interactions with neutrophils and NET induction. During gram-negative septicemia, NFAT inhibition in platelets promoted disease severity by increasing disseminated coagulation and NETosis. NFAT inhibition also partially restored coagulation ex vivo in patients with hypoactive platelets. Our results define non-transcriptional roles for NFAT that could be harnessed to address pressing clinical needs.


Asunto(s)
Plaquetas/efectos de los fármacos , Factores de Transcripción NFATC/antagonistas & inhibidores , Agregación Plaquetaria/efectos de los fármacos , Sepsis/patología , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Comunicación Celular/efectos de los fármacos , Gránulos Citoplasmáticos/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/metabolismo , Humanos , Inflamación , Ratones , Factores de Transcripción NFATC/metabolismo , Neutrófilos/metabolismo , Receptores de Trombina/metabolismo , Sepsis/metabolismo
3.
J Enzyme Inhib Med Chem ; 37(1): 202-210, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34894973

RESUMEN

Calcineurin (CN) controls the immune response by regulating nuclear factor of activated T cells (NFAT). Inhibition of CN function is an effective treatment for immune diseases. The PVIVIT peptide is an artificial peptide based on the NFAT-PxIxIT motif, which exhibits stronger binding to CN. A bioactive peptide (named pep4) that inhibits the CN/NFAT interaction was designed. Pep4 contains a segment of A238L as the linker and the LxVP motif and PVIVIT motif as CN binding sites. Pep4 has strong binding capacity to CN and inhibits CN activity competitively. 11-arginine-modified pep4 (11 R-pep4) inhibits the nuclear translocation of NFAT and reduces the expression of IL-2. 11 R-pep4 improves the pathological characteristics of asthmatic mice to a certain extent. The above results indicated that pep4 is a high-affinity CN inhibitor. These findings will contribute to the discovery of new CN inhibitors and promising immunosuppressive drugs.


Asunto(s)
Asma/tratamiento farmacológico , Calcineurina/metabolismo , Factores de Transcripción NFATC/antagonistas & inhibidores , Péptidos/farmacología , Animales , Asma/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Factores de Transcripción NFATC/metabolismo , Péptidos/síntesis química , Péptidos/química , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
4.
Int Immunopharmacol ; 101(Pt A): 108177, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34626872

RESUMEN

Osteoporosis, characterized by bone loss and microstructure damage, occurs when osteoclast activity outstrips osteoblast activity. Natural compounds with inhibitory effect on osteoclast differentiation and function have been evidenced to protect from osteoporosis. After multiple compounds screening, 12-deoxyphorbol 13-acetate (DPA) was found to decline RANKL-induced osteoclastogenesis dose-dependently by attenuating activities of NFATc1 and c-Fos, followed by decreasing the level of osteoclast function-associated genes and proteins including Acp5, V-ATPase-d2 and CTSK. Mechanistically, we found that DPA suppressing RANKL-induced downstream signaling pathways, including MAPK signaling pathway and calcium oscillations. Furthermore, the in vivo efficacy of DPA was further confirmed in an OVX-induced osteoporosis mice model. Collectively, the results in our presentation reveal that DPA might be a promising compound to manage osteoporosis.


Asunto(s)
Factores de Transcripción NFATC/antagonistas & inhibidores , Osteoporosis/tratamiento farmacológico , Ésteres del Forbol/farmacología , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Factores de Transcripción NFATC/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Osteogénesis/efectos de los fármacos , Osteogénesis/inmunología , Osteoporosis/inmunología , Ésteres del Forbol/uso terapéutico , Células RAW 264.7
5.
FASEB J ; 35(10): e21951, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34551141

RESUMEN

Deciphering the molecular and cellular processes involved in foam cell formation is critical for us to understand the pathogenesis of atherosclerosis. Nuclear factor of activated T cells (NFAT) is a transcription factor originally identified as a key player in the differentiation of T cells and maturation of immune system. Nowadays it has been brought into attention that NFAT also regulates multiple pathophysiological processes and targeted intervention in NFAT may be effective in the treatment of some cardiovascular diseases. However, whether NFAT is involved in foam cell formation remains elusive. NFAT in human monocyte-derived macrophage was activated by ox-LDL and translocated from the cytoplasm to the nucleus. NFAT then directly bound to peroxisome proliferator-activated receptor γ (PPARγ) in the nucleus and negatively regulated its transcriptional activity. NFATc2 knockdown or NFAT inhibitor 11R-VIVIT increased cholesterol efflux (by activating PPARγ-LXRα-ABCA1 cascade) and reduced the uptake of modified lipoprotein (in a PPARγ-independent way) in macrophage, thus prevented foam cell formation. Besides, 11R-VIVIT also exerted a protective role in the development of atherosclerosis in western diet-fed ApoE-/- mice. These results suggest NFAT inhibition as a potential therapeutic strategy in atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Dieta/efectos adversos , Células Espumosas/citología , Factores de Transcripción NFATC/antagonistas & inhibidores , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Núcleo Celular/metabolismo , Colesterol/metabolismo , Humanos , Lipoproteínas LDL/farmacología , Macrófagos , Masculino , Ratones , Factores de Transcripción NFATC/metabolismo , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo
6.
Acta Pharmacol Sin ; 42(11): 1860-1874, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34363007

RESUMEN

Glioblastoma multiforme (GBM) is the most common and malignant type of primary brain tumor, and 95% of patients die within 2 years after diagnosis. In this study, aiming to overcome chemoresistance to the first-line drug temozolomide (TMZ), we carried out research to discover a novel alternative drug targeting the oncogenic NFAT signaling pathway for GBM therapy. To accelerate the drug's clinical application, we took advantage of a drug repurposing strategy to identify novel NFAT signaling pathway inhibitors. After screening a set of 93 FDA-approved drugs with simple structures, we identified pimavanserin tartrate (PIM), an effective 5-HT2A receptor inverse agonist used for the treatment of Parkinson's disease-associated psychiatric symptoms, as having the most potent inhibitory activity against the NFAT signaling pathway. Further study revealed that PIM suppressed STIM1 puncta formation to inhibit store-operated calcium entry (SOCE) and subsequent NFAT activity. In cellula, PIM significantly suppressed the proliferation, migration, division, and motility of U87 glioblastoma cells, induced G1/S phase arrest and promoted apoptosis. In vivo, the growth of subcutaneous and orthotopic glioblastoma xenografts was markedly suppressed by PIM. Unbiased omics studies revealed the novel molecular mechanism of PIM's antitumor activity, which included suppression of the ATR/CDK2/E2F axis, MYC, and AuroraA/B signaling. Interestingly, the genes upregulated by PIM were largely associated with cholesterol homeostasis, which may contribute to PIM's side effects and should be given more attention. Our study identified store-operated calcium channels as novel targets of PIM and was the first to systematically highlight the therapeutic potential of pimavanserin tartrate for glioblastoma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Inhibidores de la Calcineurina/farmacología , Señalización del Calcio/efectos de los fármacos , Glioblastoma/metabolismo , Factores de Transcripción NFATC/metabolismo , Piperidinas/farmacología , Urea/análogos & derivados , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Calcineurina/metabolismo , Inhibidores de la Calcineurina/uso terapéutico , Señalización del Calcio/fisiología , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Glioblastoma/tratamiento farmacológico , Células HeLa , Humanos , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factores de Transcripción NFATC/antagonistas & inhibidores , Piperidinas/uso terapéutico , Urea/farmacología , Urea/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
7.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166238, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34343639

RESUMEN

Chronic low-grade retinal inflammation is an essential contributor to the pathogenesis of diabetic retinopathy (DR). It is characterized by increased retinal cell expression and secretion of a variety of inflammatory cytokines; among these, IL-1ß has the reputation of being a major driver of cytokine-induced inflammation. IL-1ß and other cytokines drive inflammatory changes that cause damage to retinal cells, leading to the hallmark vascular lesions of DR; these include increased leukocyte adherence, vascular permeability, and capillary cell death. Nuclear factor of activated T-cells (NFAT) is a transcriptional regulator of inflammatory cytokines and adhesion molecules and is expressed in retinal cells. Consequently, it may influence multiple pathogenic steps early in DR. We investigated the NFAT-dependency of IL-1ß-induced inflammation in human Müller cells (hMC) and human retinal microvascular endothelial cells (hRMEC). Our results show that an NFAT inhibitor, Inhibitor of NFAT-Calcineurin Association-6 (INCA-6), decreased IL-1ß-induced expression of IL-1ß and TNFα in hMC, while having no effect on VEGF, CCL2, or CCL5 expression. We also demonstrate that INCA-6 attenuated IL-1ß-induced increases of IL-1ß, TNFα, IL-6, CCL2, and CCL5 (inflammatory cytokines and chemokines), and ICAM-1 and E-selectin (leukocyte adhesion molecules) expression in hRMEC. INCA-6 similarly inhibited IL-1ß-induced increases in leukocyte adhesion in both hRMEC monolayers in vitro and an acute model of retinal inflammation in vivo. Finally, INCA-6 rescued IL-1ß-induced permeability in both hRMEC monolayers in vitro and an acute model of retinal inflammation in vivo. Taken together, these data demonstrate the potential of NFAT inhibition to mitigate retinal inflammation secondary to diabetes.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Interleucina-1beta/genética , Factores de Transcripción NFATC/genética , Vasculitis Retiniana/tratamiento farmacológico , Inhibidores de la Calcineurina/farmacología , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL5/genética , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Selectina E/genética , Células Endoteliales/efectos de los fármacos , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/patología , Molécula 1 de Adhesión Intercelular/genética , Interleucina-1beta/farmacología , Factores de Transcripción NFATC/antagonistas & inhibidores , Retina/efectos de los fármacos , Retina/patología , Vasculitis Retiniana/genética , Vasculitis Retiniana/parasitología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/patología , Factor de Necrosis Tumoral alfa/genética , Factor A de Crecimiento Endotelial Vascular/genética
8.
Bioorg Med Chem Lett ; 48: 128235, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34216746

RESUMEN

Vomifoliol, a natural sesquiterpene compound, is a secondary metabolite isolated from the mangrove plant Ceriops tagal. The present study aimed to determine the immunosuppressive effects and underlying mechanisms of vomifoliol on Jurkat cells in vitro. The results show that vomifoliol significantly inhibited calcineurin (CN) at concentrations resulting in relatively low cytotoxicity. Moreover, vomifoliol was found to exert an inhibitory effect on phorbol 12-myristate 13-acetate (PMA)/ ionomycin (Io) -induced Jurkat cells and the dephosphorylation of NFAT1. In addition, it reduced the expression of IL-2. Based on these results, we concluded that vomifoliol may inhibit the immune response of Jurkat cells, and vomifoliol may use CN as the target enzyme to inhibit NFAT signaling pathway. Therefore, vomifoliol may be promising as a low-toxic natural immunosuppressant.


Asunto(s)
Butanoles/farmacología , Ciclohexanonas/farmacología , Factores de Transcripción NFATC/antagonistas & inhibidores , Rhizophoraceae/química , Butanoles/química , Butanoles/aislamiento & purificación , Ciclohexanonas/química , Ciclohexanonas/aislamiento & purificación , Relación Dosis-Respuesta a Droga , Humanos , Células Jurkat , Estructura Molecular , Factores de Transcripción NFATC/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
9.
Aging Cell ; 20(7): e13416, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34117818

RESUMEN

Inhibition of the protein phosphatase calcineurin (CN) ameliorates pathophysiologic and cognitive changes in aging rodents and mice with aging-related Alzheimer's disease (AD)-like pathology. However, concerns over adverse effects have slowed the transition of common CN-inhibiting drugs to the clinic for the treatment of AD and AD-related disorders. Targeting substrates of CN, like the nuclear factor of activated T cells (NFATs), has been suggested as an alternative, safer approach to CN inhibitors. However, small chemical inhibitors of NFATs have only rarely been described. Here, we investigate a newly developed neuroprotective hydroxyquinoline derivative (Q134R) that suppresses NFAT signaling, without inhibiting CN activity. Q134R partially inhibited NFAT activity in primary rat astrocytes, but did not prevent CN-mediated dephosphorylation of a non-NFAT target, either in vivo, or in vitro. Acute (≤1 week) oral delivery of Q134R to APP/PS1 (12 months old) or wild-type mice (3-4 months old) infused with oligomeric Aß peptides led to improved Y maze performance. Chronic (≥3 months) oral delivery of Q134R appeared to be safe, and, in fact, promoted survival in wild-type (WT) mice when given for many months beyond middle age. Finally, chronic delivery of Q134R to APP/PS1 mice during the early stages of amyloid pathology (i.e., between 6 and 9 months) tended to reduce signs of glial reactivity, prevented the upregulation of astrocytic NFAT4, and ameliorated deficits in synaptic strength and plasticity, without noticeably altering parenchymal Aß plaque pathology. The results suggest that Q134R is a promising drug for treating AD and aging-related disorders.


Asunto(s)
Enfermedad de Alzheimer/genética , Factores de Transcripción NFATC/antagonistas & inhibidores , Placa Amiloide/fisiopatología , Animales , Modelos Animales de Enfermedad , Ratones
10.
Int J Mol Sci ; 22(5)2021 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-33800389

RESUMEN

Nuclear factor of activated T cells (NFAT), which is the pharmacological target of immunosuppressants cyclosporine and tacrolimus, has been shown to play an important role not only in T cells (immune system), from which their name is derived, but also in many biological events. Therefore, functional and/or structural abnormalities of NFAT are linked to the pathogenesis of diseases in various organs. The NFAT protein family consists of five isoforms, and each isoform performs diverse functions and has unique expression patterns in the target tissues. This diversity has made it difficult to obtain ideal pharmacological output for immunosuppressants that inhibit the activity of almost all NFAT family members, causing serious and wide-ranging side effects. Moreover, it remains unclear whether isoform-selective NFAT regulation can be achieved by targeting the structural differences among NFAT isoforms and whether this strategy can lead to the development of better drugs than the existing ones. This review summarizes the role of the NFAT family members in biological events, including the development of various diseases, as well as the usefulness of and problems associated with NFAT-targeting therapies, including those dependent on current immunosuppressants. Finally, we propose a novel therapeutic strategy based on the molecular mechanisms that enable selective regulation of specific NFAT isoforms.


Asunto(s)
Ciclosporina/uso terapéutico , Inmunosupresores/uso terapéutico , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/inmunología , Linfocitos T/inmunología , Tacrolimus/uso terapéutico , Animales , Humanos , Isoformas de Proteínas
11.
Int Immunopharmacol ; 96: 107619, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33831806

RESUMEN

Osteoporosis is a metabolic bone-loss disease characterized by abnormally excessive osteoclast formation and bone resorption. Identification of natural medicines that can inhibit osteoclastogenesis, bone resorption, and receptor activator of nuclear factor-κB ligand (RANKL)-induced signaling is necessary for improved treatment of osteoporosis. In this study, hinokitiol, a tropolone-related compound extracted from the heart wood of several cupressaceous plants, was found to inhibit RANKL-induced osteoclast formation and bone resorption in vitro. Hinokitiol inhibited early activation of the ERK, p38, and JNK-MAPK pathways, thereby suppressing the activity and expression of downstream factors (c-Jun, c-Fos, and NFATC1). Consistent with the above in vitro findings, hinokitiol treatment protected against ovariectomy-induced bone loss in vivo. Collectively, our results imply that hinokitiol can potentially serve as an effective agent for treating osteoclast-induced osteoporosis.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/prevención & control , Monoterpenos/farmacología , Osteogénesis/efectos de los fármacos , Osteoporosis/metabolismo , Osteoporosis/prevención & control , Tropolona/análogos & derivados , Actinas/antagonistas & inhibidores , Animales , Conservadores de la Densidad Ósea/uso terapéutico , Resorción Ósea/diagnóstico por imagen , Resorción Ósea/etiología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Monoterpenos/uso terapéutico , Factores de Transcripción NFATC/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Osteogénesis/genética , Ovariectomía/efectos adversos , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-jun/antagonistas & inhibidores , Ligando RANK/toxicidad , Factor de Transcripción AP-1/antagonistas & inhibidores , Tropolona/farmacología , Tropolona/uso terapéutico
12.
J Hepatol ; 74(3): 550-559, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33039404

RESUMEN

BACKGROUND & AIMS: The nuclear factor of activated T-cells (NFAT) plays an important role in immune responses by regulating the expression of inflammatory genes. However, it is not known whether NFAT plays any role in the bile acid (BA)-induced hepatic inflammatory response. Thus, we aimed to examine the functional role of NFATc3 in cholestatic liver injury in mice and humans. METHODS: Gene and protein expression and cellular localization were assessed in primary hepatocyte cultures (mouse and human) and cholestatic liver tissues (murine models and patients with primary biliary cholangitis [PBC] or primary sclerosing cholangitis [PSC]) by quantitative PCR, western blot and immunohistochemistry. Specific NFAT inhibitors were used in vivo and in vitro. Gene reporter assays and ChIP-PCR were used to determine promoter activity. RESULTS: NFAT isoforms c1 and c3 were expressed in human and mouse hepatocytes. When treated with cholestatic levels of BAs, nuclear translocation of NFATc3 was increased in both human and mouse hepatocytes and was associated with elevated mRNA levels of IL-8, CXCL2, and CXCL10 in these cells. Blocking NFAT activation with pathway-specific inhibitors or knocking down Nfatc3 expression significantly decreased BA-driven induction of these cytokines in mouse hepatocytes. Nuclear expression of NFATc3/Nfatc3 protein was increased in cholestatic livers, both in mouse models (bile duct ligation or Abcb4-/- mice) and in patients with PBC and PSC in association with elevated tissue levels of Cxcl2 (mice) or IL-8 (humans). Gene reporter assays and ChIP-PCR demonstrated that the NFAT response element in the IL-8 promoter played a key role in BA-induced human IL-8 expression. Finally, blocking NFAT activation in vivo in Abcb4-/- mice reduced cholestatic liver injury. CONCLUSIONS: NFAT plays an important role in BA-stimulated hepatic cytokine expression in cholestasis. Blocking hepatic NFAT activation may reduce cholestatic liver injury in humans. LAY SUMMARY: Bile acid induces liver injury by stimulating the expression of inflammatory genes in hepatocytes through activation of the transcription factor NFAT. Blocking this activation in vitro (in hepatocyte cultures) and in vivo (in cholestatic mice) decreased the expression of inflammatory genes and reduced liver injury.


Asunto(s)
Colangitis Esclerosante/metabolismo , Citocinas/metabolismo , Cirrosis Hepática Biliar/metabolismo , Hígado/metabolismo , Factores de Transcripción NFATC/metabolismo , Transducción de Señal/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hepatocitos/metabolismo , Humanos , Cirrosis Hepática Biliar/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/genética , Pirazoles/farmacología , Pirazoles/uso terapéutico , Transducción de Señal/genética , Resultado del Tratamiento , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
13.
Mol Med Rep ; 23(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33179105

RESUMEN

The reconstruction of pulmonary vascular structure caused by the proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) is the central link in the formation of pulmonary arterial hypertension (PAH). Platelet­derived growth factor (PDGF) can regulate the proliferation and migration of PASMCs. At the same time, nuclear factor of activated T cells (NFATs) plays an important role in the development of PAH. To the best of our knowledge, there are no reports yet regarding whether PDGF regulates NFATc2 to increase the proliferation of PASMCs. The present study aimed to investigate whether PDGF affects the proliferation and migration of PASMCs by regulating NFAT, and to study the pathogenesis of PAH. PASMCs were treated with recombinant PDGF; Cell Counting Kit­8 and clone formation experiments showed that PDGF enhanced the cell viability and proliferation of PASMCs. Cell cycle distribution and molecular markers related to cell proliferation (cyclin D1, CDK4 and Proliferating Cell Nuclear Antigen) were detected by flow cytometry, and the results indicated that PDGF promoted the division of PAMSCs. The scratch migration and Transwell migration assays showed that the migratory ability of PASMCs was enhanced following PDGF treatment. Changes in NFATs (NFATc1­5) after PDGF treatment were evaluated by reverse transcription­quantitative PCR and western blotting; NFATc2 showed the most significant results. Finally, PDGF­treated cells were treated with an NFAT pathway inhibitor, cyclosporin A, or a small interfering RNA targeting NFATc2, and changes in cell proliferation and migration were evaluated to assess the role of NFATc2 in PDGF­induced cell proliferation and migration. In conclusion, PDGF may regulate PASMC proliferation and migration by regulating the expression of NFAT, further leading to the occurrence of PAH. It is proposed that NFATc2 could be used as a potential target for PAH treatment.


Asunto(s)
Miocitos del Músculo Liso/citología , Factores de Transcripción NFATC/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Arteria Pulmonar/citología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ciclosporina/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/genética , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , ARN Interferente Pequeño/farmacología , Ratas
14.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255852

RESUMEN

Macrophages are mononuclear cells that become osteoclasts (OCs) in the presence of two cytokines, macrophage colony-stimulating factor (M-CSF), and receptor activator of NF-κB ligand (RANKL). RANKL binding to its specific receptor RANK leads to OCs differentiation mainly by nuclear factor of activated T-cells cytoplasmic 1 (NFATc1). In our previous study, the analysis of the protein network in NFATc1-knockdown cells, using the Ingenuity Pathway Analysis (IPA), showed a link between NFATc1 and Mitogen-activated protein kinase kinase (MEK)-extracellular receptor kinase (ERK) signaling pathway. Therefore, this study aimed to extend our knowledge of the relationship between NFATc1 and the ERK. Here, we demonstrate that delayed ERK1/2 phosphorylation in pre-OC RANKL-induced depends on NFATc1. Indeed, the knockdown of NFATc1 reduced the phosphorylation of ERK1/2 (60%) and the pharmacological inhibition of the ERK1/2 kinase activity impairs the expression of NFATc1 without preventing its translocation into the nucleus. Furthermore, silencing of NFATc1 significantly reduced RANKL-induced migration (p < 0.01), and most pre-OCs are still mononuclear after 48 h (80 ± 5%), despite the presence of actin rings. On the other hand, the inhibitors FR180204 and PD98059 significantly reduced RANKL-induced cell migration (p < 0.01), leading to a reduction in the number of multinucleated cells. Finally, we suggest that long-lasting ERK activity depends on NFATc1 induction and is likely linked to cell migration, fusion, and OC differentiation.


Asunto(s)
Resorción Ósea/genética , Movimiento Celular/efectos de los fármacos , Factores de Transcripción NFATC/genética , Ligando RANK/genética , Animales , Resorción Ósea/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Movimiento Celular/genética , Flavonoides/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Factores de Transcripción NFATC/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Pirazoles/farmacología , Piridazinas/farmacología , Células RAW 264.7
15.
J Am Heart Assoc ; 9(16): e016419, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32805187

RESUMEN

Background The development of pathological cardiac hypertrophy involves the coordination of a series of transcription activators and repressors, while their interplay to trigger pathological gene reprogramming remains unclear. NULP1 (nuclear localized protein 1) is a member of the basic helix-loop-helix family of transcription factors and its biological functions in pathological cardiac hypertrophy are barely understood. Methods and Results Immunoblot and immunostaining analyses showed that NULP1 expression was consistently reduced in the failing hearts of patients and hypertrophic mouse hearts and rat cardiomyocytes. Nulp1 knockout exacerbates aortic banding-induced cardiac hypertrophy pathology, which was significantly blunted by transgenic overexpression of Nulp1. Signal pathway screening revealed the nuclear factor of activated T cells (NFAT) pathway to be dramatically suppressed by NULP1. Coimmunoprecipitation showed that NULP1 directly interacted with the topologically associating domain of NFAT3 via its C-terminal region, which was sufficient to suppress NFAT3 transcriptional activity. Inactivation of the NFAT pathway by VIVIT peptides in vivo rescued the aggravated pathogenesis of cardiac hypertrophy resulting from Nulp1 deficiency. Conclusions NULP1 is an endogenous suppressor of NFAT3 signaling under hypertrophic stress and thus negatively regulates the pathogenesis of cardiac hypertrophy. Targeting overactivated NFAT by NULP1 may be a novel therapeutic strategy for the treatment of pathological cardiac hypertrophy and heart failure.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cardiomegalia/metabolismo , Factores de Transcripción NFATC/metabolismo , Proteínas Represoras/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/genética , Cardiomegalia/terapia , Ecocardiografía , Eliminación de Gen , Humanos , Inmunoprecipitación/métodos , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/genética , Oligopéptidos/farmacología , Monoéster Fosfórico Hidrolasas/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transcripción Genética
16.
Biomed Pharmacother ; 130: 110523, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32702632

RESUMEN

Bone disorders such as osteoporosis, Paget's disease of the bone, osteogenesis imperfecta, are caused by the uncoordinated action of osteoclasts and osteoblasts. Inhibiting osteoclastogenesis and suppressing the resorptive function of osteoclasts might become a gold standard strategy for treating this kind of disease. Glucagon-like peptide-1 (GLP-1) and its receptor agonist have been reported to have protective effects on bone. Little is known about the effect of GLP-1 on osteoclasts. Therefore, we investigated the effects of liraglutide, a GLP-1 receptor agonist, on murine bone marrow-derived macrophage (BMM) and RAW264.7 preosteoclast differentiation and explored the potential cellular basis of its action. In this study, we confirmed the presence of GLP-1 receptor (GLP-1R) on BMMs and RAW264.7 cells and demonstrated that GLP-1R might be important for osteoclastogenesis by increasing the expression of osteoclastogenic biomarkers after GLP-1R knockdown. In addition, we found that liraglutide treatment of both BMMs and RAW264.7 cells could inhibit osteoclast formation and bone resorption. Mechanistically, Western blotting and RT-PCR showed that liraglutide inhibited the NF-κB and MAPK signalling pathways, ultimately inhibiting the expression of nuclear factor of activated T cells (NFATc1). In addition, knocking down GLP-1R reversed the inhibitory effect of liraglutide on NF-κB/MAPK-NFATc1. Overall, these results indicated a potential therapeutic effect of liraglutide on bone disorders.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/agonistas , Liraglutida/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Resorción Ósea/prevención & control , Diferenciación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Receptor del Péptido 1 Similar al Glucagón/genética , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/biosíntesis , Ligando RANK/antagonistas & inhibidores , Células RAW 264.7
17.
J Hepatol ; 73(6): 1333-1346, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32717288

RESUMEN

BACKGROUND & AIMS: The nuclear factor of activated T-cells (NFAT) family was first recognised to play an important role in the differentiation of T cells, but has since been shown to regulate multiple pathophysiological processes. However, whether it is involved in the pathogenesis of non-alcoholic steatohepatitis (NASH) remains unknown. METHODS: Hepatic NFATc expression and localisation were analysed in C57BL/6 mice on a methionine-choline-deficient diet, as well as in samples from non-alcoholic fatty liver disease patients. Gain- or loss-of-function approaches were used to investigate the role of NFATc4 in NASH. RESULTS: NFATc4 translocates from the cytoplasm to the nucleus in hepatocytes of both humans and rodents with NASH. NFATc4 knockdown resulted in decreased hepatic steatosis, inflammation, and fibrosis during NASH progression. Mechanistically, we found that activated NFATc4 directly bound to peroxisome proliferator-activated receptor α (PPARα) in the nucleus and negatively regulated its transcriptional activity, thereby impairing the hepatic fatty acid oxidation pathway and increasing lipid deposition in the liver. Moreover, NFATc4 activation increased the production and secretion of osteopontin (OPN) from hepatocytes, which subsequently enhanced the macrophage-mediated inflammatory response and hepatic stellate cell-mediated fibrosis progression via paracrine signalling. CONCLUSIONS: Hepatic NFATc4 activation accelerates the progression of NASH by suppressing PPARα signalling and increasing OPN expression. Genetic or pharmacological inhibition of NFATc4 may have potential for future therapy of NASH. LAY SUMMARY: NFATc4 is activated in the non-alcoholic steatohepatitis of mice and patients. Inhibition of NFATc4 activation alleviates lipid deposition, inflammatory response, and fibrosis progression in the liver.


Asunto(s)
Cirrosis Hepática , Factores de Transcripción NFATC/metabolismo , Enfermedad del Hígado Graso no Alcohólico , Osteopontina/metabolismo , PPAR alfa/metabolismo , Animales , Progresión de la Enfermedad , Descubrimiento de Drogas , Perfilación de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/antagonistas & inhibidores , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Comunicación Paracrina , Transducción de Señal
18.
Cancer Med ; 9(15): 5558-5569, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32515123

RESUMEN

OSW-1 is a natural compound extracted from the bulbs of Ornithogalum saundersiae in 1992. It has been shown strong antitumor activities in various cancer cells. However, the effects of OSW-1 on tumor growth and metastasis in breast cancer are still poorly understood. In our research, we showed that OSW-1 had a strong anticancer effect on breast cancer cells, but lower toxicity to normal cells. Accordingly, it also revealed significant inhibition of tumor growth by OSW-1 in xenograft model. In addition, we performed Annexin V/PI-labeled flow cytometric assay and TUNEL assay and showed that OSW-1 inhibited tumor growth by inducing apoptosis. Furthermore, we carried out transwell assays and found that OSW-1 significantly repressed the migratory and invasive capabilities of triple-negative breast cancer (TNBC) cells via mediating epithelial-mesenchymal transition. Besides, OSW-1 also could inhibit metastasis in an orthotopic model and resulted in a longer survival compared with control group. Finally, we performed RNA-sequencing and cellular functions to investigate the molecular mechanism of how OSW-1 inhibits TNBC, and identified NFATc2 may as a pivotal factor for OSW-1-mediated effects on cell death, tumor growth, invasion, and migration.


Asunto(s)
Colestenonas/farmacología , Factores de Transcripción NFATC/metabolismo , Ornithogalum/química , Saponinas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones SCID , Factores de Transcripción NFATC/antagonistas & inhibidores , Metástasis de la Neoplasia
19.
Cells ; 9(5)2020 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-32397368

RESUMEN

The tumor suppressor p53 is believed to be the mostly studied molecule in modern biomedical research. Although p53 interacts with hundreds of molecules to exert its biological functions, there are only a few modulators regulating its expression and function, with murine double minute 2 (MDM2) playing a key role in this regard. MDM2 also contributes to malignant transformation and cancer development through p53-dependent and -independent mechanisms. There is an increasing interest in developing MDM2 inhibitors for cancer prevention and therapy. We recently demonstrated that the nuclear factor of activated T cells 1 (NFAT1) activates MDM2 expression. NFAT1 regulates several cellular functions in cancer cells, such as cell proliferation, migration, invasion, angiogenesis, and drug resistance. Both NFAT isoforms and MDM2 are activated and overexpressed in several cancer subtypes. In addition, a positive correlation exists between NFAT1 and MDM2 in tumor tissues. Our recent clinical study has demonstrated that high expression levels of NFAT1 and MDM2 are independent predictors of a poor prognosis in patients with hepatocellular carcinoma. Thus, inhibition of the NFAT1-MDM2 pathway appears to be a novel potential therapeutic strategy for cancer. In this review, we summarize the potential oncogenic roles of MDM2 and NFAT1 in cancer cells and discuss the efforts of discovery and the development of several newly identified MDM2 and NFAT1 inhibitors, focusing on their potent in vitro and in vivo anticancer activities. This review also highlights strategies and future directions, including the need to focus on the development of more specific and effective NFAT1-MDM2 dual inhibitors for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Factores de Transcripción NFATC/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Animales , Descubrimiento de Drogas , Humanos , Terapia Molecular Dirigida , Factores de Transcripción NFATC/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo
20.
Gastroenterology ; 159(3): 1036-1050.e8, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32445858

RESUMEN

BACKGROUND & AIMS: Calcineurin is a ubiquitously expressed central Ca2+-responsive signaling molecule that mediates acute pancreatitis, but little is known about its effects. We compared the effects of calcineurin expression by hematopoietic cells vs pancreas in mouse models of pancreatitis and pancreatitis-associated lung inflammation. METHODS: We performed studies with mice with hematopoietic-specific or pancreas-specific deletion of protein phosphatase 3, regulatory subunit B, alpha isoform (PPP3R1, also called CNB1), in mice with deletion of CNB1 (Cnb1UBC△/△) and in the corresponding controls for each deletion of CNB1. Acute pancreatitis was induced in mice by administration of caerulein or high-pressure infusion of radiocontrast into biliopancreatic ducts; some mice were also given intraductal infusions of an adeno-associated virus vector that expressed nuclear factor of activated T -cells (NFAT)-luciferase into pancreas. Pancreas, bone marrow, liver, kidney, heart, and lung were collected and analyzed by histopathology, immunohistochemistry, and immunoblots; levels of cytokines were measured in serum. Mouse and human primary pancreatic acinar cells were transfected with a vector that expressed NFAT-luciferase and incubated with an agent that blocks interaction of NFAT with calcineurin; cells were analyzed by immunofluorescence. Calcineurin-mediated neutrophil chemotaxis and reactive oxygen species production were measured in neutrophils from mice. RESULTS: Mice with hematopoietic-specific deletion of CNB1 developed the same level of local pancreatic inflammation as control mice after administration of caerulein or infusion of radiocontrast into biliopancreatic ducts. Cnb1UBC△/△ mice or mice with pancreas-specific deletion of CNB1 developed less severe pancreatitis and reduced pancreatic inflammation after administration of caerulein or infusion of radiocontrast into biliopancreatic ducts compared with control mice. NFAT was activated in pancreas of Swiss Webster mice given caerulein or infusions of radiocontrast into biliopancreatic ducts. Blocking the interaction between calcineurin and NFAT did not reduce pancreatic acinar cell necrosis in response to caerulein or infusions of radiocontrast. Mice with hematopoietic-specific deletion of CNB1 (but not mice with pancreas-specific deletion of CNB1) had reduced infiltration of lung tissues by neutrophils. Neutrophil chemotaxis and production of reactive oxygen species were decreased after incubation with a calcineurin inhibitor. CONCLUSIONS: Hematopoietic and neutrophil expression of calcineurin promotes pancreatitis-associated lung inflammation, whereas pancreatic calcineurin promotes local pancreatic inflammation. The findings indicate that the protective effects of blocking or deleting calcineurin on pancreatitis are mediated by the source of its expression. This information should be used in the development of strategies to inhibit calcineurin for the prevention of pancreatitis and pancreatitis-associated lung inflammation.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Inhibidores de la Calcineurina/uso terapéutico , Calcineurina/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Musculares/metabolismo , Pancreatitis/inmunología , Células Acinares/metabolismo , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/prevención & control , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Calcineurina/genética , Calcineurina/inmunología , Proteínas de Unión al Calcio/genética , Células Cultivadas , Ceruletida/administración & dosificación , Ceruletida/toxicidad , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Páncreas/citología , Páncreas/inmunología , Páncreas/metabolismo , Pancreatitis/inducido químicamente , Pancreatitis/complicaciones , Pancreatitis/tratamiento farmacológico , Cultivo Primario de Células
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...