Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 388
Filtrar
1.
Biochem Biophys Res Commun ; 715: 150004, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38678784

RESUMEN

Megakaryopoiesis and platelet production is a complex process that is underpotential regulation at multiple stages. Many long non-coding RNAs (lncRNAs) are distributed in hematopoietic stem cells and platelets. lncRNAs may play important roles as key epigenetic regulators in megakaryocyte differentiation and proplatelet formation. lncRNA NORAD can affect cell ploidy by sequestering PUMILIO proteins, although its direct effect on megakaryocyte differentiation and thrombopoiesis is still unknown. In this study, we demonstrate NORAD RNA is highly expressed in the cytoplasm during megakaryocyte differentiation. Interestingly, we identified for the first time that NORAD has a strong inhibitory effect on megakaryocyte differentiation and proplatelet formation from cultured megakaryocytes. DUSP6/ERK1/2 pathway is activated in response to NORAD knockdown during megakaryocytopoiesis, which is achieved by sequestering PUM2 proteins. Finally, compared with the wild-type control mice, NORAD knockout mice show a faster platelet recovery after severe thrombocytopenia induced by 6 Gy total body irradiation. These findings demonstrate lncRNA NORAD has a key role in regulating megakaryocyte differentiation and thrombopoiesis, which provides a promising molecular target for the treatment of platelet-related diseases such as severe thrombocytopenia.


Asunto(s)
Plaquetas , Diferenciación Celular , Fosfatasa 6 de Especificidad Dual , Megacariocitos , Ratones Noqueados , ARN Largo no Codificante , Trombopoyesis , Megacariocitos/metabolismo , Megacariocitos/citología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Animales , Trombopoyesis/genética , Plaquetas/metabolismo , Ratones , Fosfatasa 6 de Especificidad Dual/metabolismo , Fosfatasa 6 de Especificidad Dual/genética , Sistema de Señalización de MAP Quinasas , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patología , Humanos , Ratones Endogámicos C57BL , Células Cultivadas
2.
J Neurosci ; 44(5)2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38296649

RESUMEN

The mesolimbic dopamine system is a crucial component of reward and reinforcement processing, including the psychotropic effects of drugs of abuse such as cocaine. Drugs of abuse can activate intracellular signaling cascades that engender long-term molecular changes to brain reward circuitry, which can promote further drug use. However, gaps remain about how the activity of these signaling pathways, such as ERK1/2 signaling, can affect cocaine-induced neurochemical plasticity and cocaine-associated behaviors specifically within dopaminergic cells. To enable specific modulation of ERK1/2 signaling in dopaminergic neurons of the ventral tegmental area, we utilize a viral construct that Cre dependently expresses Map kinase phosphatase 3 (MKP3) to reduce the activity of ERK1/2, in combination with transgenic rats that express Cre in tyrosine hydroxylase (TH)-positive cells. Following viral transfection, we found an increase in the surface expression of the dopamine transporter (DAT), a protein associated with the regulation of dopamine signaling, dopamine transmission, and cocaine-associated behavior. We found that inactivation of ERK1/2 reduced post-translational phosphorylation of the DAT, attenuated the ability of cocaine to inhibit the DAT, and decreased motivation for cocaine without affecting associative learning as tested by conditioned place preference. Together, these results indicate that ERK1/2 signaling plays a critical role in shaping the dopamine response to cocaine and may provide additional insights into the function of dopaminergic neurons. Further, these findings lay important groundwork toward the assessment of how signaling pathways and their downstream effectors influence dopamine transmission and could ultimately provide therapeutic targets for treating cocaine use disorders.


Asunto(s)
Cocaína , Dopamina , Ratas , Animales , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Motivación , Sistema de Señalización de MAP Quinasas , Fosfatasa 6 de Especificidad Dual/metabolismo , Cocaína/farmacología , Área Tegmental Ventral/fisiología , Recompensa , Ratas Transgénicas
3.
Int J Mol Sci ; 24(21)2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37958545

RESUMEN

Drug-induced liver injury (DILI) is a widespread and harmful disease, and is closely linked to acute endoplasmic reticulum (ER) stress. Previous reports have shown that acute ER stress can suppress hepatic gluconeogenesis and even leads to hypoglycemia. However, the mechanism is still unclear. MAPK phosphatase 3 (MKP-3) is a positive regulator for gluconeogenesis. Thus, this study was conducted to investigate the role of MKP-3 in the suppression of gluconeogenesis by acute ER stress, as well as the regulatory role of acute ER stress on the expression of MKP-3. Results showed that acute ER stress induced by tunicamycin significantly suppressed gluconeogenesis in both hepatocytes and mouse liver, reduced glucose production level in hepatocytes, and decreased fasting blood glucose level in mice. Additionally, the protein level of MKP-3 was reduced by acute ER stress in both hepatocytes and mouse liver. Mkp-3 deficiency eliminated the inhibitory effect of acute ER stress on gluconeogenesis in hepatocytes. Moreover, the reduction effect of acute ER stress on blood glucose level and hepatic glucose 6-phosphatase (G6pc) expression was not observed in the liver-specific Mkp-3 knockout mice. Furthermore, activation of protein kinase R-like ER kinase (PERK) decreased the MKP-3 protein level, while inactivation of PERK abolished the reduction effect of acute ER stress on the MKP-3 protein level in hepatocytes. Taken together, our study suggested that acute ER stress could suppress hepatic gluconeogenesis by stimulating MKP-3 degradation via PERK, at least partially. Thus, MKP-3 might be a therapeutic target for DILI-related hypoglycemia.


Asunto(s)
Fosfatasa 6 de Especificidad Dual , Gluconeogénesis , Hipoglucemia , Animales , Ratones , Glucemia/metabolismo , Estrés del Retículo Endoplásmico , Hepatocitos/metabolismo , Hipoglucemia/metabolismo , Hígado/metabolismo , Ratones Noqueados , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfatasa 6 de Especificidad Dual/metabolismo
4.
Chemosphere ; 344: 140358, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37797900

RESUMEN

Particulate matter (PM), one of the most serious air contaminants, could easily pass through the airway and deposit at the deep alveoli. Thus, it might trigger respiratory diseases like inflammation, asthma and lung cancer on human. Long non-coding RNAs (lncRNAs) are considered as important regulator in promotion and progression of diverse cancers. However, the molecular mechanism of lncRNAs mediating PM-induced lung carcinogenesis remains unclear. In this study, we established a 16HBE malignant transformed cell induced by PM (Cells were treated with 20 µg/ml PM, which named PM-T cells) and explored the roles and mechanisms of lncRNAs in the malignant transformation induced by PM. Compared with 16HBE cells, various biological functions were changed in PM-T cells, such as cell proliferation, migration, cell cycle and apoptosis. LncRNA SPRY4-IT1 was significant down-regulated expression and associated with these biological effects. Our results showed that lncRNA SPRY4-IT1 overexpression reversed these functional changes mentioned above. The further studies indicated that lncRNA SPRY4-IT1 involved in PM-induced cell transformation by modulating Chk1 expression via negative regulation of DUSP6-ERK1/2. In conclusion, our studies suggested that lncRNA SPRY4-IT1 played the role as a tumor suppressor gene and might mediate 16HBE cells malignant transformation induced by PM through regulating DUSP6-ERK1/2-Chk1 signaling pathway.


Asunto(s)
ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Línea Celular Tumoral , Sistema de Señalización de MAP Quinasas , Regulación Neoplásica de la Expresión Génica , Transducción de Señal , Proliferación Celular/genética , Movimiento Celular , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo
5.
Arch Esp Urol ; 76(6): 445-453, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37681336

RESUMEN

BACKGROUND: Nucleolar prominence is a biomarker of prostate cancer (CaP), and the nucleolar protein block of proliferation 1 (BOP1) participates in the development of CaP, which has great significance for CaP therapy. Thus, this study explored the mechanism of BOP1 in CaP development. METHODS: BOP1 expression levels in the tumor tissues of CaP patients and in PC3 tumor cells were determined. The viability, apoptosis rate of PC3 cells, and apoptosis-related proteins levels were determined to explore the effect of BOP1 on tumor-cell growth in vitro. BOP1 function in the metastasis of PC3 cells was further assessed by Transwell experiment. We also studied the influence of BOP1 on the expression of mitogen-activated protein kinase (MAPK) pathway-related proteins and investigated the regulatory effect of BOP1 on dual-specificity phosphatase 6 (DUSP6). RESULTS: BOP1 expression was upregulated in the tumor tissues and PC3 cells of CaP patients. BOP1 knockout reduced the activity of PC3 cells and induced apoptosis, significantly inhibiting the metastasis of PC3 cells. DUSP6 was overexpressed in tumor tissues and PC3 cells. BOP1 knockout inhibited DUSP6 expression and the MAPK pathway. DUSP6 overexpression reversed the inhibition of BOP1 siRNA (si-BOP1) on PC3 cells and the activated MAPK signaling pathway. CONCLUSIONS: This finding demonstrated that BOP1 promoted CaP progression by regulating the DUSP6/MAPK pathway.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos , Neoplasias de la Próstata , Masculino , Humanos , Neoplasias de la Próstata/genética , Transducción de Señal , Proliferación Celular , Fosfatasa 6 de Especificidad Dual/genética , Proteínas de Unión al ARN
6.
Arch. esp. urol. (Ed. impr.) ; 76(6): 445-453, 28 aug. 2023. ilus, graf
Artículo en Inglés | IBECS | ID: ibc-224897

RESUMEN

Background: Nucleolar prominence is a biomarker of prostate cancer (CaP), and the nucleolar protein block of proliferation 1 (BOP1) participates in the development of CaP, which has great significance for CaP therapy. Thus, this study explored the mechanism of BOP1 in CaP development. Methods: BOP1 expression levels in the tumor tissues of CaP patients and in PC3 tumor cells were determined. The viability, apoptosis rate of PC3 cells, and apoptosis-related proteins levels were determined to explore the effect of BOP1 on tumor-cell growth in vitro. BOP1 function in the metastasis of PC3 cells was further assessed by Transwell experiment. We also studied the influence of BOP1 on the expression of mitogen-activated protein kinase (MAPK) pathway-related proteins and investigated the regulatory effect of BOP1 on dual-specificity phosphatase 6 (DUSP6). Results: BOP1 expression was upregulated in the tumor tissues and PC3 cells of CaP patients. BOP1 knockout reduced the activity of PC3 cells and induced apoptosis, significantly inhibiting the metastasis of PC3 cells. DUSP6 was overexpressed in tumor tissues and PC3 cells. BOP1 knockout inhibited DUSP6 expression and the MAPK pathway. DUSP6 overexpression reversed the inhibition of BOP1 siRNA (si-BOP1) on PC3 cells and the activated MAPK signaling pathway. Conclusions: This finding demonstrated that BOP1 promoted CaP progression by regulating the DUSP6/MAPK pathway (AU)


Asunto(s)
Humanos , Masculino , Neoplasias de la Próstata/sangre , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosfatasa 6 de Especificidad Dual/metabolismo , Transducción de Señal , Proliferación Celular , Biomarcadores de Tumor , Apoptosis
7.
J Assist Reprod Genet ; 40(7): 1597-1610, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37300650

RESUMEN

PURPOSE: Dysregulated behaviors of trophoblast cells leading to defective placentation are considered the main cause of preeclampsia (PE). Abnormal miRNA expression profiles have been observed in PE placental tissue, indicating the significant role of miRNAs in PE development. This study aimed to investigate the expression of miR-101-5p in PE placental tissue and its biological functions. METHODS: The expression of miR-101-5p in placental tissue was detected by quantitative real-time PCR (qRT-PCR). The localization of miR-101-5p in term placental tissue and decidual tissue was determined by the fluorescence in situ hybridization (FISH)-immunofluorescence (IF) double labeling assay. The effect of miR-101-5p on the migration, invasion, proliferation, and apoptosis of the HTR8/SVneo trophoblast cells was investigated. Online databases combined with transcriptomics were used to identify potential target genes and related pathways of miR-101-5p. Finally, the interaction between miR-101-5p and the target gene was verified by qRT-PCT, WB, dual-luciferase reporter assay, and rescue experiments. RESULTS: The study found that miR-101-5p was upregulated in PE placental tissue compared to normal controls and was mainly located in various trophoblast cell subtypes in placental and decidual tissues. Overexpression of miR-101-5p impaired the migration and invasion of HTR8/SVneo cells. DUSP6 was identified as a potential downstream target of miR-101-5p. The expression of miR-101-5p was negatively correlated with DUSP6 expression in HTR8/SVneo cells, and miR-101-5p directly bound to the 3' UTR region of DUSP6. DUSP6 upregulation rescued the migratory and invasive abilities of HTR8/SVneo cells in the presence of miR-101-5p overexpression. Additionally, miR-101-5p downregulated DUSP6, resulting in enhanced ERK1/2 phosphorylation. CONCLUSION: This study revealed that miR-101-5p inhibits the migration and invasion of HTR8/SVneo cells by regulating the DUSP6-ERK1/2 axis, providing a new molecular mechanism for the pathogenesis of PE.


Asunto(s)
MicroARNs , Preeclampsia , Humanos , Embarazo , Femenino , Placenta/metabolismo , Trofoblastos/metabolismo , Preeclampsia/patología , Hibridación Fluorescente in Situ , Sistema de Señalización de MAP Quinasas/genética , Línea Celular , MicroARNs/genética , MicroARNs/metabolismo , Proliferación Celular/genética , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo
8.
BMB Rep ; 56(9): 508-513, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37291053

RESUMEN

The phytochemical quercetin has gained attention for its antiinflammatory and anti-tumorigenic properties in various types of cancer. Tumorigenesis involves the aberrant regulation of kinase/phosphatase, highlighting the importance of maintaining homeostasis. Dual Specificity Phosphatase (DUSP) plays a crucial role in controlling the phosphorylation of ERK. The current study aimed to clone the DUSP5 promoter, and investigate its transcriptional activity in the presence of quercetin. The results revealed that quercetin-induced DUSP5 expression is associated with the serum response factor (SRF) binding site located in the DUSP5 promoter. The deletion of this site abolished the luciferase activity induced by quercetin, indicating its vital role in quercetin-induced DUSP5 expression. SRF protein is a transcription factor that potentially contributes to quercetin-induced DUSP5 expression at the transcriptional level. Additionally, quercetin enhanced SRF binding activity without changing its expression. These findings provide evidence of how quercetin affects anti-cancer activity in colorectal tumorigenesis by inducing SRF transcription factor activity, thereby increasing DUSP5 expression at the transcriptional level. This study highlights the importance of investigating the molecular mechanisms underlying the anti-cancer properties of quercetin, and suggests its potential use in cancer therapy. [BMB Reports 2023; 56(9): 508-513].


Asunto(s)
Quercetina , Factor de Respuesta Sérica , Humanos , Quercetina/farmacología , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Fosforilación , Carcinogénesis , Fosfatasa 6 de Especificidad Dual/metabolismo
9.
Sci Rep ; 13(1): 5683, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-37029196

RESUMEN

Cultured human pluripotent stem cells (hPSCs) grow as colonies that require breakdown into small clumps for further propagation. Although cell death mechanism by single-cell dissociation of hPSCs has been well defined, how hPSCs respond to the deadly stimulus and recover the original status remains unclear. Here we show that dissociation of hPSCs immediately activates ERK, which subsequently activates RSK and induces DUSP6, an ERK-specific phosphatase. Although the activation is transient, DUSP6 expression persists days after passaging. DUSP6 depletion using the CRISPR/Cas9 system reveals that DUSP6 suppresses the ERK activity over the long term. Elevated ERK activity by DUSP6 depletion increases both viability of hPSCs after single-cell dissociation and differentiation propensity towards mesoderm and endoderm lineages. These findings provide new insights into how hPSCs respond to dissociation in order to maintain pluripotency.


Asunto(s)
Células Madre Pluripotentes , Transducción de Señal , Humanos , Retroalimentación , Diferenciación Celular , Muerte Celular , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo
10.
Int J Mol Sci ; 24(6)2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36982663

RESUMEN

We investigated the expression and biological function of retinoic acid inducible gene I (RIG-I) in esophageal squamous cell carcinoma (ESCC). Materials and methods: An immunohistochemical analysis was performed on 86 pairs of tumor tissue and adjacent normal tissue samples of patients with ESCC. We generated RIG-I-overexpressing ESCC cell lines KYSE70 and KYSE450, and RIG-I- knockdown cell lines KYSE150 and KYSE510. Cell viability, migration and invasion, radioresistance, DNA damage, and cell cycle were evaluated using CCK-8, wound-healing and transwell assay, colony formation, immunofluorescence, and flow cytometry and Western blotting, respectively. RNA sequencing was performed to determine the differential gene expression between controls and RIG-I knockdown. Tumor growth and radioresistance were assessed in nude mice using xenograft models. RIG-I expression was higher in ESCC tissues compared with that in matched non-tumor tissues. RIG-I overexpressing cells had a higher proliferation rate than RIG-I knockdown cells. Moreover, the knockdown of RIG-I slowed migration and invasion rates, whereas the overexpression of RIG-I accelerated migration and invasion rates. RIG-I overexpression induced radioresistance and G2/M phase arrest and reduced DNA damage after exposure to ionizing radiations compared with controls; however, it silenced the RIG-I enhanced radiosensitivity and DNA damage, and reduced the G2/M phase arrest. RNA sequencing revealed that the downstream genes DUSP6 and RIG-I had the same biological function; silencing DUSP6 can reduce the radioresistance caused by the overexpression of RIG-I. RIG-I knockdown depleted tumor growth in vivo, and radiation exposure effectively delayed the growth of xenograft tumors compared with the control group. RIG-I enhances the progression and radioresistance of ESCC; therefore, it may be a new potential target for ESCC-targeted therapy.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Animales , Humanos , Ratones , Carcinogénesis/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Fosfatasa 6 de Especificidad Dual/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/radioterapia , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/genética , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , Receptores de Ácido Retinoico/metabolismo
11.
Ren Fail ; 45(1): 2173950, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36794657

RESUMEN

The renal interstitial fibrosis contributes to the progression and deterioration of diabetic nephropathy (DN). Long noncoding RNA taurine-up-regulated gene 1 (TUG1) in kidneys may be down-regulated by hyperglycemia. We aim to explore its role in tubular fibrosis caused by high glucose and the possible target genes of TUG1. In this study, a streptozocin-induced accelerated DN mouse model and a high glucose-stimulated HK-2 cells model was established to evaluate TUG1 expression. Potential targets of TUG1 were analyzed by online tools and confirmed by luciferase assay. A rescue experiment and gene silencing assay were used to investigate whether TUG1 plays its regulation role via miR-145-5p/dual-specificity phosphatase 6 (DUSP6) in HK2 cells. The effects of TUG1 on inflammation and fibrosis in high glucose treated tubular cells were evaluated by in vitro study, as well as in vivo DN mice model through AAV-TUG1 delivery. Results showed TUG1was downregulated in HK2 cells incubated with high glucose while miR-145-5p was upregulated. Overexpression of TUG1 alleviated renal injury by suppressing inflammation and fibrosis in vivo. Overexpression of TUG1 inhibited HK-2 cell fibrosis and relieved the inflammation. A mechanism study demonstrated that TUG1 directly sponged to miR-145-5p, and DUSP6 was identified as a target downstream of miR-145-5p. In addition, miR-145-5 overexpression and DUSP6 inhibition countervailed the impacts of TUG1. Our findings revealed that TUG1 overexpression alleviates kidney injury in DN mice and decreases the inflammatory response and fibrosis of high glucose-stimulated HK-2 cells via miR-145-5p/DUSP6 axis.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Fosfatasa 6 de Especificidad Dual , MicroARNs , ARN Largo no Codificante , Animales , Ratones , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Fosfatasa 6 de Especificidad Dual/metabolismo , Fibrosis , Glucosa , Inflamación , MicroARNs/genética , MicroARNs/metabolismo
12.
Cancer Lett ; 558: 216092, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36806557

RESUMEN

Despite many advances in treatment over the past few years, the poor 5-year survival rate and high recurrence rate of gastric cancer (GC) remain unsatisfactory. As the most abundant epigenetic modification in the eukaryotic mRNA, N6-methyladenosine (m6A) methylation participates in tumor progression and tissue development. During tumor progression, DNA damage repair mechanisms can be reprogrammed to give new growth advantages on tumor clones whose genomic integrity is disturbed. Here we detected the elevated SUV39H2 expression in GC tissues and cell lines. Functionally, SUV39H2 promoted GC proliferation and inhibited apoptosis in vitro and in vivo. Mechanistically, METTL3-mediated m6A modification promotes mRNA stability of SUV39H2 in an IGF2BP2 dependent manner, resulting in upregulated mRNA expression of SUV39H2. As a histone methyltransferase, SUV39H2 was verified to increase the phosphorylation level of ATM through transcriptional repression of DUSP6, thereby promoting HRR and ultimately inhibiting GC chemosensitivity to cisplatin. Collectively, these results indicate the specific mechanism of m6A-modified SUV39H2 as a histone methyltransferase promoting HRR to inhibit the chemosensitivity of GC. SUV39H2 is expected to become a key target in the precision targeted therapy of GC.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patología , Represión Epigenética , Línea Celular Tumoral , Recombinación Homóloga , Histona Metiltransferasas/genética , ARN Mensajero , Metiltransferasas/metabolismo , Proteínas de Unión al ARN/genética , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo , N-Metiltransferasa de Histona-Lisina/genética
13.
Nat Cancer ; 4(1): 108-127, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36581736

RESUMEN

Myeloproliferative neoplasms (MPNs) exhibit a propensity for transformation to secondary acute myeloid leukemia (sAML), for which the underlying mechanisms remain poorly understood, resulting in limited treatment options and dismal clinical outcomes. Here, we performed single-cell RNA sequencing on serial MPN and sAML patient stem and progenitor cells, identifying aberrantly increased expression of DUSP6 underlying disease transformation. Pharmacologic dual-specificity phosphatase (DUSP)6 targeting led to inhibition of S6 and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling while also reducing inflammatory cytokine production. DUSP6 perturbation further inhibited ribosomal S6 kinase (RSK)1, which we identified as a second indispensable candidate associated with poor clinical outcome. Ectopic expression of DUSP6 mediated JAK2-inhibitor resistance and exacerbated disease severity in patient-derived xenograft (PDX) models. Contrastingly, DUSP6 inhibition potently suppressed disease development across Jak2V617F and MPLW515L MPN mouse models and sAML PDXs without inducing toxicity in healthy controls. These findings underscore DUSP6 in driving disease transformation and highlight the DUSP6-RSK1 axis as a vulnerable, druggable pathway in myeloid malignancies.


Asunto(s)
Leucemia Mieloide Aguda , Trastornos Mieloproliferativos , Animales , Ratones , Humanos , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Transducción de Señal/genética , Quinasas Janus/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Fosfatasa 6 de Especificidad Dual/metabolismo
14.
Reprod Sci ; 30(6): 1782-1788, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36474130

RESUMEN

Ovulatory disorders are a major cause of infertility in humans as well as economically important species. In physiological conditions, the LH surge induces the expression of epidermal growth factor (EGF)-like ligands that activate the EGR receptor (EGFR) and subsequently the mitogen-activated protein kinase (MAPK) pathway. The magnitude and duration of MAPK phosphorylation are regulated by dual-specificity phosphatases (DUSPs). Besides this well-known cascade, other signaling pathways such as the Hippo pathway modulate the ovulatory cascade and are reported to crosstalk with MAPK signaling. Here, we tested the hypothesis that LH and the Hippo pathway regulate DUSP expression in bovine pre-ovulatory granulosa cells. The abundance of DUSP6 mRNA but not DUSP1 was decreased by LH (P < 0.05). Cells were then pre-treated (1 h) with two inhibitors of Hippo signaling, verteporfin (1 µM) or peptide-17 (25 µM), before exposure for 6 h to LH or to EGF. Treatment with verteporfin increased DUSP1 mRNA levels (P < 0.05) in the presence or absence of EGF or LH and treatment with peptide-17 increased DUSP6 and not DUSP1 mRNA abundance. These data indicate a differential regulation of DUSP1 and DUSP6 mRNA by the Hippo pathway in pre-ovulatory granulosa cells, which suggests a complex control of MAPK signaling around ovulation.


Asunto(s)
Fosfatasas de Especificidad Dual , Factor de Crecimiento Epidérmico , Femenino , Humanos , Animales , Bovinos , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Vía de Señalización Hippo , Verteporfina , ARN Mensajero , Células de la Granulosa/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatasa 6 de Especificidad Dual/metabolismo
16.
Dis Model Mech ; 16(5)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36478044

RESUMEN

Acute myocardial infarction (MI) results in loss of cardiomyocytes and abnormal cardiac remodeling with severe inflammation and fibrosis. However, how cardiac repair can be achieved by timely resolution of inflammation and cardiac fibrosis remains incompletely understood. Our previous findings have shown that dual-specificity phosphatase 6 (DUSP6) is a regeneration repressor from zebrafish to rats. In this study, we found that intravenous administration of the DUSP6 inhibitor (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI) improved heart function and reduced cardiac fibrosis in MI rats. Mechanistic analysis revealed that BCI attenuated macrophage inflammation through NF-κB and p38 signaling, independent of DUSP6 inhibition, leading to the downregulation of various cytokines and chemokines. In addition, BCI suppressed differentiation-related signaling pathways and decreased bone-marrow cell differentiation into macrophages through inhibiting DUSP6. Furthermore, intramyocardial injection of poly (D, L-lactic-co-glycolic acid)-loaded BCI after MI had a notable effect on cardiac repair. In summary, BCI improves heart function and reduces abnormal cardiac remodeling by inhibiting macrophage formation and inflammation post-MI, thus providing a promising pro-drug candidate for the treatment of MI and related heart diseases. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Infarto del Miocardio , Animales , Ratas , Fosfatasa 6 de Especificidad Dual , Fibrosis , Infarto del Miocardio/complicaciones , Infarto del Miocardio/tratamiento farmacológico , Remodelación Ventricular
17.
Aging (Albany NY) ; 14(23): 9679-9698, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36462498

RESUMEN

The most hostile form of urologic cancer, clear cell renal cell carcinoma (ccRCC), has a high fatality rate and poor prognosis due to tumor metastasis at initial presentation. The complex process driving ccRCC metastasis is still unknown, though. In this study, we demonstrate that Spindle and kinetochore-associated protein 1 (SKA1) expression is significantly upregulated in ccRCC tissues and associated with aggressive clinicopathologic characteristics. Functionally, SKA1 knockdown on ccRCC cells reduced cancer cell motility both in vivo and in vitro research. These bioactivities of SKA1 may be brought on by its specific interaction with scaffold attachment factor B, according to the proposed mechanism (SAFB), which could further depress the transcription of dual specificity phosphatase 6 (DUSP6). Our findings may provide a new way of researching SKA1-regulated tumor metastasis, and indicate that SKA1 is a prospective therapeutic target for renal carcinoma.


Asunto(s)
Carcinoma de Células Renales , Carcinoma , Neoplasias Renales , Proteínas de Unión a la Región de Fijación a la Matriz , Humanos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Fosfatasa 6 de Especificidad Dual , Neoplasias Renales/genética , Neoplasias Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Metástasis de la Neoplasia , Movimiento Celular/genética , Receptores de Estrógenos , Proteínas Asociadas a Matriz Nuclear
18.
Nat Commun ; 13(1): 6672, 2022 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-36335128

RESUMEN

Dual-specificity phosphatase 6 (DUSP6) serves a specific and conserved function on the dephosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). We previously identified Dusp6 as a regenerative repressor during zebrafish heart regeneration, therefore we propose to investigate the role of this repressor in mammalian cardiac repair. Utilizing a rat strain harboring Dusp6 nonsense mutation, rat neutrophil-cardiomyocyte co-culture, bone marrow transplanted rats and neutrophil-specific Dusp6 knockout mice, we find that Dusp6 deficiency improves cardiac outcomes by predominantly attenuating neutrophil-mediated myocardial damage in acute inflammatory phase after myocardial infarction. Mechanistically, Dusp6 is transcriptionally activated by p38-C/EBPß signaling and acts as an effector for maintaining p-p38 activity by down-regulating pERK and p38-targeting phosphatases DUSP1/DUSP16. Our findings provide robust animal models and novel insights for neutrophil-mediated cardiac damage and demonstrate the potential of DUSP6 as a therapeutic target for post-MI cardiac remodeling and other relevant inflammatory diseases.


Asunto(s)
Infarto del Miocardio , Animales , Ratones , Ratas , Fosfatasa 6 de Especificidad Dual , Ratones Noqueados , Infarto del Miocardio/genética , Miocardio , Miocitos Cardíacos , Neutrófilos
19.
Life Sci ; 309: 121040, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36208663

RESUMEN

AIMS: Several signaling events have been identified for mediating cisplatin-induced chronic inflammation and progressive renal fibrosis, but the majority of them have not yet been established as therapeutic targets. This study investigated the modulatory effects of berberine on purinergic 2X7 receptors (P2X7R) and some potential intracellular profibrogenic signaling as molecular mechanisms that could hinder renal fibrosis associated with cisplatin administration in rats. MAIN METHODS: For induction of kidney injury, rats were injected with cisplatin (1 mg/kg, i.p.) daily for two weeks. Concurrently, the rats were treated with berberine (100 or 200 mg/kg, p.o). The gene expressions of P2X7R, dual-specificity phosphatase 6 (DUSP6), and murine double-minute 2 (MDM2) were determined. The expressions of alpha smooth-muscle actin and tumor necrosis factor alpha (TNF-α) were assessed by immunohistochemical staining. Phosphorylated extracellular signal-regulated kinase 1/2, (p-ERK1/2) was evaluated by western blotting. Sirtuin 2 (SIRT2), kidney injury molecule-1, and galectin-3 were measured by enzyme-linked immunosorbent assay. The degree of renal fibrosis was assessed by microscopic examination and picrosirius red staining. KEY FINDINGS: Berberine effectively inhibited cisplatin-induced renal histopathological changes, enhanced renal function, and markedly mitigated inflammatory and fibrotic alterations as well as TNF-α protein expression. Additionally, P2X7R, p-ERK1/2, MDM2, and SIRT2 were suppressed and DUSP6 was upregulated by berberine. SIGNIFICANCE: The nephroprotective effects of berberine were mediated in part by downregulating P2X7R and modulating DUSP6-mediated inactivation of ERK1/2 as well as by suppressing SIRT2/MDM2-triggered renal fibrosis.


Asunto(s)
Berberina , Enfermedades Renales , Ratas , Ratones , Animales , Cisplatino/toxicidad , Berberina/farmacología , Sirtuina 2 , Factor de Necrosis Tumoral alfa , Fosfatasa 6 de Especificidad Dual , Proteína Quinasa 3 Activada por Mitógenos , Sistema de Señalización de MAP Quinasas , Actinas , Galectina 3 , Enfermedades Renales/inducido químicamente , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/prevención & control , Fibrosis
20.
Pathol Int ; 72(11): 529-540, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36161420

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) develops via dysplastic changes in the epithelia graded as low- and high-grade with accumulation of molecular alterations. Constitutive activation of mitogen-activated protein kinase (MAPK) contributed by attenuation of DUSP6 plays a key role in sustaining PDAC. Active MAPK induces various molecules that function as effectors to sustain PDAC. AURKA and SON are downstream effectors that contribute substantially to the proliferation and survival of PDAC cells and are potentially useful as therapeutic targets. Active MAPK also promote microRNAs that modulate the proliferation of PDAC cells and are useful as diagnostic markers. Familial pancreatic cancer kindreds in Japan show various germline mutations supposed to increase a pancreatic cancer risk. Intraductal papillary mucinous neoplasms (IPMNs) consist of dilated ducts lined by papillary neoplastic epithelia of various shapes and varying grades of atypia. Various papillae of IPMNs are classified into four subtypes that are associated with clinicopathological features, including patient prognosis. GNAS is a specific driver gene for the development of IPMN through gain-of-function mutations. Tracing of molecular alterations has elucidated the mechanism of progression of IPMN from dysplasia to carcinoma, as well as one type of papillae. Intraductal tubulopapillary neoplasms belong to a distinct class of pancreatic neoplasms.


Asunto(s)
Carcinogénesis , Carcinoma Ductal Pancreático , Neoplasias Intraductales Pancreáticas , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/patología , Neoplasias Intraductales Pancreáticas/genética , Neoplasias Intraductales Pancreáticas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Carcinogénesis/genética , Carcinogénesis/patología , Fosfatasa 6 de Especificidad Dual/genética , Aurora Quinasa A/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...