Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Cell Mol Life Sci ; 81(1): 33, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38214819

RESUMEN

P38γ (MAPK12) is predominantly expressed in triple negative breast cancer cells (TNBC) and induces stem cell (CSC) expansion resulting in decreased survival of the patients due to metastasis. Abundance of G-rich sequences at MAPK12 promoter implied the functional probability to reverse tumorigenesis, though the formation of G-Quadruplex (G4) structures at MAPK12 promoter is elusive. Here, we identified two evolutionary consensus adjacent G4 motifs upstream of the MAPK12 promoter, forming parallel G4 structures. They exist in an equilibria between G4 and duplex, regulated by the binding turnover of Sp1 and Nucleolin that bind to these G4 motifs and regulate MAPK12 transcriptional homeostasis. To underscore the gene-regulatory functions of G4 motifs, we employed CRISPR-Cas9 system to eliminate G4s from TNBC cells and synthesized a naphthalene diimide (NDI) derivative (TGS24) which shows high-affinity binding to MAPK12-G4 and inhibits MAPK12 transcription. Deletion of G4 motifs and NDI compound interfere with the recruitment of the transcription factors, inhibiting MAPK12 expression in cancer cells. The molecular basis of NDI-induced G4 transcriptional regulation was analysed by RNA-seq analyses, which revealed that MAPK12-G4 inhibits oncogenic RAS transformation and trans-activation of NANOG. MAPK12-G4 also reduces CD44High/CD24Low population in TNBC cells and downregulates internal stem cell markers, arresting the stemness properties of cancer cells.


Asunto(s)
G-Cuádruplex , Proteína Quinasa 12 Activada por Mitógenos , Neoplasias de la Mama Triple Negativas , Humanos , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Neoplasias de la Mama Triple Negativas/genética , Proteína Quinasa 12 Activada por Mitógenos/genética
2.
Brain ; 147(5): 1871-1886, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38128553

RESUMEN

Multiple sclerosis is a chronic inflammatory disease in which disability results from the disruption of myelin and axons. During the initial stages of the disease, injured myelin is replaced by mature myelinating oligodendrocytes that differentiate from oligodendrocyte precursor cells. However, myelin repair fails in secondary and chronic progressive stages of the disease and with ageing, as the environment becomes progressively more hostile. This may be attributable to inhibitory molecules in the multiple sclerosis environment including activation of the p38MAPK family of kinases. We explored oligodendrocyte precursor cell differentiation and myelin repair using animals with conditional ablation of p38MAPKγ from oligodendrocyte precursors. We found that p38γMAPK ablation accelerated oligodendrocyte precursor cell differentiation and myelination. This resulted in an increase in both the total number of oligodendrocytes and the migration of progenitors ex vivo and faster remyelination in the cuprizone model of demyelination/remyelination. Consistent with its role as an inhibitor of myelination, p38γMAPK was significantly downregulated as oligodendrocyte precursor cells matured into oligodendrocytes. Notably, p38γMAPK was enriched in multiple sclerosis lesions from patients. Oligodendrocyte progenitors expressed high levels of p38γMAPK in areas of failed remyelination but did not express detectable levels of p38γMAPK in areas where remyelination was apparent. Our data suggest that p38γ could be targeted to improve myelin repair in multiple sclerosis.


Asunto(s)
Esclerosis Múltiple , Vaina de Mielina , Oligodendroglía , Remielinización , Animales , Remielinización/fisiología , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Ratones , Oligodendroglía/metabolismo , Oligodendroglía/patología , Humanos , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/genética , Diferenciación Celular/fisiología , Cuprizona/toxicidad , Ratones Endogámicos C57BL , Masculino , Femenino , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/patología , Ratones Transgénicos
3.
Elife ; 122023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37458356

RESUMEN

Evidence implicating p38γ and p38δ (p38γ/p38δ) in inflammation are mainly based on experiments using Mapk12/Mapk13-deficient (p38γ/δKO) mice, which show low levels of TPL2, the kinase upstream of MKK1-ERK1/2 in myeloid cells. This could obscure p38γ/p38δ roles, since TPL2 is essential for regulating inflammation. Here, we generated a Mapk12D171A/D171A/Mapk13-/- (p38γ/δKIKO) mouse, expressing kinase-inactive p38γ and lacking p38δ. This mouse exhibited normal TPL2 levels, making it an excellent tool to elucidate specific p38γ/p38δ functions. p38γ/δKIKO mice showed a reduced inflammatory response and less susceptibility to lipopolysaccharide (LPS)-induced septic shock and Candida albicans infection than wild-type (WT) mice. Gene expression analyses in LPS-activated wild-type and p38γ/δKIKO macrophages revealed that p38γ/p38δ-regulated numerous genes implicated in innate immune response. Additionally, phospho-proteomic analyses and in vitro kinase assays showed that the transcription factor myocyte enhancer factor-2D (MEF2D) was phosphorylated at Ser444 via p38γ/p38δ. Mutation of MEF2D Ser444 to the non-phosphorylatable residue Ala increased its transcriptional activity and the expression of Nos2 and Il1b mRNA. These results suggest that p38γ/p38δ govern innate immune responses by regulating MEF2D phosphorylation and transcriptional activity.


Asunto(s)
Lipopolisacáridos , Proteína Quinasa 13 Activada por Mitógenos , Animales , Ratones , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Proteómica , Inmunidad Innata , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Inflamación
4.
Cancer Gene Ther ; 30(9): 1181-1189, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37248432

RESUMEN

Gastrointestinal cancers are a leading cause of cancer morbidity and mortality worldwide with 4.2 million new cases and 3.2 million deaths estimated in 2020. Despite the advances in primary and adjuvant therapies, patients still develop distant metastases and require novel therapies. Mitogen­activated protein kinase (MAPK) cascades are crucial signaling pathways that regulate many cellular processes, including proliferation, differentiation, apoptosis, stress responses and cancer development. p38 Mitogen Activated Protein Kinases (p38 MAPKs) includes four isoforms: p38α (MAPK14), p38ß (MAPK11), p38γ (MAPK12), and p38δ (MAPK13). p38 MAPK was first identified as a stress response protein kinase that phosphorylates different transcriptional factors. Dysregulation of p38 pathways, in particular p38γ, are associated with cancer development, metastasis, autophagy and tumor microenvironment. In this article, we provide an overview of p38 and p38γ with respect to gastrointestinal cancers. Furthermore, targeting p38γ is also discussed as a potential therapy for gastrointestinal cancers.


Asunto(s)
Neoplasias Gastrointestinales , Proteína Quinasa 11 Activada por Mitógenos , Humanos , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Transducción de Señal , Neoplasias Gastrointestinales/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Microambiente Tumoral
5.
BMC Plant Biol ; 23(1): 53, 2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36694135

RESUMEN

BACKGROUND: Many data suggest that the sucrose non-fermenting 1-related kinases 2 (SnRK2s) are very important to abiotic stress for plants. In rice, these kinases are known as osmotic stress/ABA-activated protein kinases (SAPKs). Osmotic stress/ABA-activated protein kinase 3 (OsSAPK3) is a member of SnRK2II in rice, but its function is still unclear. RESULTS: The expression of OsSAPK3 was up regulated by drought, NaCl, PEG and ABA. OsSAPK3 mutated seedings (sapk3-1 and sapk3-2) showed reduced hypersensitivity to exogenous ABA. In addition, under drought conditions, sapk3-1 and sapk3-2 showed more intolerance to drought, including decreased survival rate, increased water loss rate, increased stomatal conductance and significantly decreased expression levels of SLAC1 and SLAC7. Physiological and metabolic analyses showed that OsSAPK3 might play an important role in drought stress signaling pathway by affecting osmotic adjustment and osmolytes, ROS detoxification and expression of ABA dependent and independent dehydration-responsive genes. All gronomic traits analyses demonstrated that OsSAPK3 could improve rice yield by affecting the regulation of tiller numbers and grain size. CONCLUSION: OsSAPK3 plays an important role in both ABA-dependent and ABA-independent drought stress responses. More interestingly, OsSAPK3 could improve rice yield by indirectly regulating tiller number and grain size. These findings provide new insight for the development of drought-resistant rice.


Asunto(s)
Resistencia a la Sequía , Oryza , Oryza/metabolismo , Proteínas de Plantas/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Grano Comestible/genética , Sequías , Estrés Fisiológico/genética , Plantas Modificadas Genéticamente/genética , Regulación de la Expresión Génica de las Plantas , Ácido Abscísico/metabolismo
6.
Proc Natl Acad Sci U S A ; 119(35): e2204752119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994673

RESUMEN

p38γ and p38δ (p38γ/p38δ) regulate inflammation, in part by controlling tumor progression locus 2 (TPL2) expression in myeloid cells. Here, we demonstrate that TPL2 protein levels are dramatically reduced in p38γ/p38δ-deficient (p38γ/δ-/-) cells and tissues without affecting TPL2 messenger ribonucleic acid (mRNA) expression. We show that p38γ/p38δ posttranscriptionally regulates the TPL2 amount at two different levels. p38γ/p38δ interacts with the TPL2/A20 Binding Inhibitor of NF-κB2 (ABIN2)/Nuclear Factor κB1p105 (NF-κB1p105) complex, increasing TPL2 protein stability. Additionally, p38γ/p38δ regulates TPL2 mRNA translation by modulating the repressor function of TPL2 3' Untranslated region (UTR) mediated by its association with aconitase-1 (ACO1). ACO1 overexpression in wild-type cells increases the translational repression induced by TPL2 3'UTR and severely decreases TPL2 protein levels. p38δ binds to ACO1, and p38δ expression in p38γ/δ-/- cells fully restores TPL2 protein to wild-type levels by reducing the translational repression of TPL2 mRNA. This study reveals a unique mechanism of posttranscriptional regulation of TPL2 expression, which given its central role in innate immune response, likely has great relevance in physiopathology.


Asunto(s)
Aconitato Hidratasa , Quinasas Quinasa Quinasa PAM , Proteína Quinasa 12 Activada por Mitógenos , Proteína Quinasa 13 Activada por Mitógenos , Aconitato Hidratasa/genética , Aconitato Hidratasa/metabolismo , Regulación de la Expresión Génica , Inmunidad Innata , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , ARN Mensajero/genética
7.
BMC Endocr Disord ; 21(1): 235, 2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34814904

RESUMEN

BACKGROUND: Prolactinoma is a functional pituitary adenoma that secretes excessive prolactin. Dopamine agonists (DAs) such as bromocriptine (BRC) are the first-line treatment for prolactinomas, but the resistance rate is increasing year by year, creating a clinical challenge. Therefore, it is urgent to explore the molecular mechanism of bromocriptine resistance in prolactinomas. Activation of the P38 MAPK pathway affects multidrug resistance in tumours. Our previous studies have demonstrated that inhibiting MAPK14 can suppress the occurrence of prolactinoma, but the role of MAPK11/12/13/14 (p38 MAPK) signalling in dopamine agonist-resistant prolactinomas is still unclear. METHODS: A prolactinoma rat model was established to determine the effect of bromocriptine on MAPK11/12/13/14 signalling. DA-resistant GH3 cells and DA-sensitive MMQ cells were used, and the role of MAPK11/12/13/14 in bromocriptine-resistant prolactinomas was preliminarily verified by western blot, RT-qPCR, ELISA, flow cytometry and CCK-8 experiments. The effects of MAPK11 or MAPK14 on bromocriptine-resistant prolactinomas were further verified by siRNA transfection experiments. RESULTS: Bromocriptine was used to treat rat prolactinoma by upregulating DRD2 expression and downregulating the expression level of MAPK11/12/13/14 in vivo experiments. The in vitro experiments showed that GH3 cells are resistant to bromocriptine and that MMQ cells are sensitive to bromocriptine. Bromocriptine could significantly reduce the expression of MAPK12 and MAPK13 in GH3 cells and MMQ cells. Bromocriptine could significantly reduce the expression of MAPK11, MAPK14, NF-κB p65 and Bcl2 in MMQ but had no effect on MAPK11, MAPK14, NF-κB p65 and Bcl2 in GH3 cells. In addition, knockdown of MAPK11 and MAPK14 in GH3 cells by siRNA transfection reversed the resistance of GH3 cells to bromocriptine, and haloperidol (HAL) blocked the inhibitory effect of bromocriptine on MAPK14, MAPK11, and PRL in MMQ cells. Our findings show that MAPK11 and MAPK14 proteins are involved in bromocriptine resistance in prolactinomas. CONCLUSION: Bromocriptine reduces the expression of MAPK11/12/13/14 in prolactinomas, and MAPK11 and MAPK14 are involved in bromocriptine resistance in prolactinomas by regulating apoptosis. Reducing the expression of MAPK11 or MAPK14 can reverse bromocriptine resistance in prolactinomas.


Asunto(s)
Agonistas de Dopamina/uso terapéutico , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/enzimología , Prolactinoma/tratamiento farmacológico , Prolactinoma/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Apoptosis , Bromocriptina/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Estradiol/administración & dosificación , Estradiol/análogos & derivados , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Prolactina/genética , Prolactinoma/inducido químicamente , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/genética , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/genética
8.
Am J Physiol Heart Circ Physiol ; 319(4): H775-H786, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32822209

RESUMEN

The efficacy of an anthracycline antibiotic doxorubicin (DOX) as a chemotherapeutic agent is limited by dose-dependent cardiotoxicity. DOX is associated with activation of intracellular stress signaling pathways including p38 MAPKs. While previous studies have implicated p38 MAPK signaling in DOX-induced cardiac injury, the roles of the individual p38 isoforms, specifically, of the alternative isoforms p38γ and p38δ, remain uncharacterized. We aimed to determine the potential cardioprotective effects of p38γ and p38δ genetic deletion in mice subjected to acute DOX treatment. Male and female wild-type (WT), p38γ-/-, p38δ-/-, and p38γ-/-δ-/- mice were injected with 30 mg/kg DOX and their survival was tracked for 10 days. During this period, cardiac function was assessed by echocardiography and electrocardiography and fibrosis by Picro Sirius Red staining. Immunoblotting was performed to assess the expression of signaling proteins and markers linked to autophagy. Significantly improved survival was observed in p38δ-/- female mice post-DOX relative to WT females, but not in p38γ-/- or p38γ-/-δ-/- male or female mice. The improved survival in DOX-treated p38δ-/- females was associated with decreased fibrosis, increased cardiac output and LV diameter relative to DOX-treated WT females, and similar to saline-treated controls. Structural and echocardiographic parameters were either unchanged or worsened in all other groups. Increased autophagy, as suggested by increased LC3-II level, and decreased mammalian target of rapamycin activation was also observed in DOX-treated p38δ-/- females. p38δ plays a crucial role in promoting DOX-induced cardiotoxicity in female mice by inhibiting autophagy. Therefore, p38δ targeting could be a potential cardioprotective strategy in anthracycline chemotherapy.NEW & NOTEWORTHY This study for the first time identifies the sex-specific roles of the alternative p38γ and p38δ MAPK isoforms in promoting doxorubicin (DOX) cardiotoxicity. We show that p38δ and p38γ/δ systemic deletion was cardioprotective in female but not in male mice. Cardiac structure and function were preserved in DOX-treated p38δ-/- females and autophagy marker was increased.


Asunto(s)
Doxorrubicina , Cardiopatías/prevención & control , Proteína Quinasa 13 Activada por Mitógenos/deficiencia , Miocardio/enzimología , Animales , Autofagia/efectos de los fármacos , Cardiotoxicidad , Modelos Animales de Enfermedad , Femenino , Fibrosis , Técnicas de Inactivación de Genes , Cardiopatías/enzimología , Cardiopatías/genética , Cardiopatías/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/deficiencia , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Miocardio/patología , Factores Sexuales , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
9.
Cancer Res ; 80(16): 3251-3264, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32580961

RESUMEN

KRAS is mutated in most pancreatic ductal adenocarcinomas (PDAC) and yet remains undruggable. Here, we report that p38γ MAPK, which promotes PDAC tumorigenesis by linking KRAS signaling and aerobic glycolysis (also called the Warburg effect), is a novel therapeutic target. p38γ interacted with a glycolytic activator PFKFB3 that was dependent on mutated KRAS. KRAS transformation and overexpression of p38γ increased expression of PFKFB3 and glucose transporter GLUT2, conversely, silencing mutant KRAS, and p38γ decreased PFKFB3 and GLUT2 expression. p38γ phosphorylated PFKFB3 at S467, stabilized PFKFB3, and promoted their interaction with GLUT2. Pancreatic knockout of p38γ decreased p-PFKFB3/PFKFB3/GLUT2 protein levels, reduced aerobic glycolysis, and inhibited PDAC tumorigenesis in KPC mice. PFKFB3 and GLUT2 depended on p38γ to stimulate glycolysis and PDAC growth and p38γ required PFKFB3/S467 to promote these activities. A p38γ inhibitor cooperated with a PFKFB3 inhibitor to blunt aerobic glycolysis and PDAC growth, which was dependent on p38γ. Moreover, overexpression of p38γ, p-PFKFB3, PFKFB3, and GLUT2 in PDAC predicted poor clinical prognosis. These results indicate that p38γ links KRAS oncogene signaling and aerobic glycolysis to promote pancreatic tumorigenesis through PFKFB3 and GLUT2, and that p38γ and PFKFB3 may be targeted for therapeutic intervention in PDAC. SIGNIFICANCE: These findings show that p38γ links KRAS oncogene signaling and the Warburg effect through PFKBF3 and Glut2 to promote pancreatic tumorigenesis, which can be disrupted via inhibition of p38γ and PFKFB3.


Asunto(s)
Carcinoma Ductal Pancreático/etiología , Transportador de Glucosa de Tipo 2/metabolismo , Glucólisis , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Neoplasias Pancreáticas/etiología , Fosfofructoquinasa-2/antagonistas & inhibidores , Fosfofructoquinasa-2/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Aerobiosis , Animales , Carcinoma Ductal Pancreático/prevención & control , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Colágeno , Combinación de Medicamentos , Femenino , Técnicas de Inactivación de Genes , Silenciador del Gen , Genes ras , Técnicas de Genotipaje , Humanos , Laminina , Masculino , Ratones , Proteína Quinasa 12 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/prevención & control , Fosforilación , Pronóstico , Proteoglicanos , Proteínas Proto-Oncogénicas p21(ras)/genética
10.
FASEB J ; 33(12): 13131-13144, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31638431

RESUMEN

Despite the high and preferential expression of p38γ MAPK in the myocardium, little is known about its function in the heart. The aim of the current study was to elucidate the physiologic and biochemical roles of p38γ in the heart. Expression and subcellular localization of p38 isoforms was determined in mouse hearts. Comparisons of the cardiac function and structure of wild-type and p38γ knockout (KO) mice at baseline and after abdominal aortic banding demonstrated that KO mice developed less ventricular hypertrophy and that contractile function is better preserved. To identify potential substrates of p38γ, we generated an analog-sensitive mutant to affinity tag endogenous myocardial proteins. Among other proteins, this technique identified calpastatin as a direct p38γ substrate. Moreover, phosphorylation of calpastatin by p38γ impaired its ability to inhibit the protease, calpain. We have identified p38γ as an important determinant of the progression of pathologic cardiac hypertrophy after aortic banding in mice. In addition, we have identified calpastatin, among other substrates, as a novel direct target of p38γ that may contribute to the protection observed in p38γKO mice.-Loonat, A. A., Martin, E. D., Sarafraz-Shekary, N., Tilgner, K., Hertz, N. T., Levin, R., Shokat, K. M., Burlingame, A. L., Arabacilar, P., Uddin, S., Thomas, M., Marber, M. S., Clark, J. E. p38γ MAPK contributes to left ventricular remodeling after pathologic stress and disinhibits calpain through phosphorylation of calpastatin.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Calpaína/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Remodelación Ventricular/fisiología , Animales , Calpaína/genética , Ecocardiografía , Electroforesis en Gel de Poliacrilamida , Células HEK293 , Humanos , Inmunohistoquímica , Masculino , Ratones , Proteína Quinasa 12 Activada por Mitógenos/genética , Fosforilación , Isoformas de Proteínas , Espectrometría de Masas en Tándem , Remodelación Ventricular/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Biochem Biophys Res Commun ; 517(1): 172-179, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31349971

RESUMEN

Colorectal cancer (CRC) is a common malignancy globally causing significant cancer-related mortality. Recent studies have proposed p38gamma (p38γ) as a novel cyclin-dependent kinase (CDK)-like kinase, promoting tumorigenesis and cancer progression. The current study evaluates p38γ expression and potential role in CRC. In HT-29 cells and primary human colon cancer cells, shRNA-induced p38γ silencing or CRISPR/Cas9-mediated p38γ knockout inhibited cell growth, proliferation, and migration, and induced significant apoptosis. Conversely, ectopic overexpression of p38γ further promoted the growth, proliferation, and migration of HT-29 cells and primary colon cancer cells. Retinoblastoma (Rb) phosphorylation and cyclins (E1/A) expression were decreased by p38γ silencing or KO, but increased with p38γ overexpression. p38γ mRNA and protein levels are significantly upregulated in human colon cancer tissues, when compared to levels in surrounding colon epithelial tissues. These results demonstrate that overexpression of p38γ can promote human CRC cell progression, and identify p38γ as a novel therapeutic target.


Asunto(s)
Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Proteína Quinasa 12 Activada por Mitógenos/genética , Apoptosis , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Células HT29 , Humanos
12.
Cell Death Dis ; 10(6): 376, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092814

RESUMEN

Apoptosis and senescence are two mutually exclusive cell fate programs that can be activated by stress. The factors that instruct cells to enter into senescence or apoptosis are not fully understood, but both programs can be regulated by the stress kinase p38α. Using an inducible system that specifically activates this pathway, we show that sustained p38α activation suffices to trigger massive autophagosome formation and to enhance the basal autophagic flux. This requires the concurrent effect of increased mitochondrial reactive oxygen species production and the phosphorylation of the ULK1 kinase on Ser-555 by p38α. Moreover, we demonstrate that macroautophagy induction by p38α signaling determines that cancer cells preferentially enter senescence instead of undergoing apoptosis. In agreement with these results, we present evidence that the induction of autophagy by p38α protects cancer cells from chemotherapy-induced apoptosis by promoting senescence. Our results identify a new mechanism of p38α-regulated basal autophagy that controls the fate of cancer cells in response to stress.


Asunto(s)
Autofagia , Senescencia Celular , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Homólogo de la Proteína 1 Relacionada con la Autofagia/antagonistas & inhibidores , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Senescencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MAP Quinasa Quinasa 6/antagonistas & inhibidores , MAP Quinasa Quinasa 6/genética , MAP Quinasa Quinasa 6/metabolismo , Mitocondrias/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/deficiencia , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo , Transducción de Señal
13.
Int J Biochem Cell Biol ; 107: 6-13, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30447427

RESUMEN

BACKGROUND: The expression of p38 MAPK is high in breast cancer while its subunit p38γ had been rarely reported. We aimed to explain the effect of p38γ in breast cancer from the perspective of metabolomics. METHODS: In this study, we detected the expression of p38γ in 28 breast carcinoma and para-tumor samples. Following MDA-MB-231 cell transfection with p38γ siRNAs and pc-DNA-3.1, cell viability, apoptosis, metastasis were determined through CCK-8, the cytometry analysis, transwell assay and wound healing assay. Finally, gas chromatograph-mass spectrometer (GC-MS) was used for analysis the differential metabolites. RESULTS: The expression of p38γ was significantly up-regulated in breast cancer tissues. The transfection of si-p38γs could inhibit MDA-MB-231 cell propagation, metastasis, and induced cell apoptosis while overexpressed p38γ could promote the cell propagation, metastasis, and inhibit cell apoptosis. A total of 238 metabolites were identified and 72 of them differentially expressed in three groups (all P < 0.05, FDR < 0.05). Then the metabolites were enriched in the metabolism pathway, 85 pathways were included and 27 were significant (all P < 0.05, FDR < 0.05). CONCLUSIONS: p38γ was up-regulated in breast cancer, which exerts a great influence on the cell growth, cell mobility, invasiveness, and apoptosis of MDA-MB-231 cells and also affected the metabolism.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Metabolómica , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Apoptosis , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Proteína Quinasa 12 Activada por Mitógenos/genética , Invasividad Neoplásica
14.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3605-3617, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30251680

RESUMEN

p38γ is a member of p38 MAPK family which contains four isoforms p38α, p38ß, p38γ, and p38δ. p38γ MAPK has unique function and is less investigated. Recent studies revealed that p38γ MAPK may be involved in tumorigenesis and cancer aggressiveness. However, the underlying cellular/molecular mechanisms remain unclear. Epithelial-mesenchymal transition (EMT) is a process that epithelial cancer cells transform to facilitate the loss of epithelial features and gain of mesenchymal phenotype. EMT promotes cancer cell progression and metastasis, and is involved in the regulation of cancer stem cells (CSCs) which have self-renewal capacity and are resistant to chemotherapy and target therapy. We showed that p38γ MAPK significantly increased EMT in breast cancer cells; over-expression of p38γ MAPK enhanced EMT while its down-regulation inhibited EMT. Meanwhile, p38γ MAPK augmented CSC population while knock down of p38γ MAPK decreased CSC ratio in breast cancer cells. MicroRNA-200b (miR-200b) was down-stream of p38γ MAPK and inhibited by p38γ MAPK; miR-200b mimics blocked p38γ MAPK-induced EMT while miR-200b inhibitors promoted EMT. p38γ MAPK regulated miR-200b through inhibiting GATA3. p38γ MAPK induced GATA3 ubiquitination, leading to its proteasome-dependent degradation. Suz12, a Polycomb group protein, was down-stream of miR-200b and involved in miR-200b regulation of EMT. Thus, our study established an important role of p38γ MAPK in EMT and identified a novel signaling pathway for p38γ MAPK-mediated tumor promotion.


Asunto(s)
Transición Epitelial-Mesenquimal , Factor de Transcripción GATA3/metabolismo , MicroARNs/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Células Madre Neoplásicas/patología , Neoplasias de la Mama/patología , Carcinogénesis/genética , Regulación hacia Abajo , Femenino , Factor de Transcripción GATA3/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas/genética , Células MCF-7 , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Proteína Quinasa 12 Activada por Mitógenos/genética , Células Madre Neoplásicas/metabolismo , ARN Interferente Pequeño/metabolismo , Ubiquitinación
15.
J Cell Sci ; 131(14)2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-29930081

RESUMEN

Aggresome formation is a major strategy to enable cells to cope with proteasomal stress. Misfolded proteins are assembled into micro-aggregates and transported to the microtubule organizing center (MTOC) to form perinuclear aggresomes before their degradation through autophagy. So far, multiple factors have been identified as the activators of micro-aggregate formation, but much less is known about the regulatory mechanisms of their transport. Here, we report that proteasomal stress leads to the activation of p38 MAPK family members. Two of them, p38γ (MAPK12) and p38δ (MAPK13), are dispensable for micro-aggregate formation but are required for their targeting to the MTOC. Interestingly, p38δ promotes micro-aggregate transport by phosphorylating SQSTM1, a major scaffold protein that assembles soluble ubiquitylated proteins into micro-aggregates. Expression of the phospho-mimetic mutant of SQSTM1 in p38δ-knockout cells completely rescued their aggresome formation defects and enhanced their resistance to proteasomal stress to wild-type levels. This study reveals p38δ-mediated SQSTM1 phosphorylation as a critical signal for the targeting of micro-aggregates to the MTOC and provides direct evidence for the survival advantages associated with aggresome formation in cells under proteasomal stress.


Asunto(s)
Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Sequestosoma-1/metabolismo , Humanos , Centro Organizador de los Microtúbulos/enzimología , Centro Organizador de los Microtúbulos/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/genética , Fosforilación , Complejo de la Endopetidasa Proteasomal/genética , Agregado de Proteínas , Transporte de Proteínas , Proteína Sequestosoma-1/genética
16.
J Invest Dermatol ; 138(11): 2377-2387, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29758280

RESUMEN

Current cutaneous T-cell lymphoma (CTCL) therapies are marked by an abbreviated response, subsequent drug resistance, and poor prognosis for patients with advanced disease. An understanding of molecular regulators involved in CTCL is needed to develop effective targeted therapies. One candidate regulator is p38γ, a mitogen-activated protein kinase crucial for malignant T-cell activity and growth. p38γ gene expression is selectively increased in CTCL patient samples and cell lines but not in healthy T cells. In addition, gene silencing of p38γ reduced CTCL cell viability, showing a key role in CTCL pathogenesis. Screening p38γ inhibitors is critical for understanding the mechanism of CTCL tumorigenesis and developing therapeutic applications. We prioritized a potent p38γ inhibitor (F7, also known as PIK75) through a high-throughput kinase inhibitor screen. At nanomolar concentrations, PIK75, a multiple kinase inhibitor, selectively killed CD4+ malignant CTCL cells but spared healthy CD4+ cells; induced significant reduction of tumor size in mouse xenografts; and effectively inhibited p38γ enzymatic activity and phosphorylation of its substrate, DLGH1, in CTCL cells and mouse xenografts. Here, we report that PIK75 has a potential clinical application to serve as a scaffold molecule for the development of a more selective p38γ inhibitor.


Asunto(s)
Antineoplásicos/uso terapéutico , Linfocitos T CD4-Positivos/fisiología , Linfoma Cutáneo de Células T/tratamiento farmacológico , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Carcinogénesis , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Proteína Quinasa 12 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 12 Activada por Mitógenos/genética , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , Carga Tumoral , Células Tumorales Cultivadas , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
17.
EMBO Mol Med ; 10(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29661910

RESUMEN

Candida albicans is a frequent aetiologic agent of sepsis associated with high mortality in immunocompromised patients. Developing new antifungal therapies is a medical need due to the low efficiency and resistance to current antifungal drugs. Here, we show that p38γ and p38δ regulate the innate immune response to C. albicans We describe a new TAK1-TPL2-MKK1-ERK1/2 pathway in macrophages, which is activated by Dectin-1 engagement and positively regulated by p38γ/p38δ. In mice, p38γ/p38δ deficiency protects against C. albicans infection by increasing ROS and iNOS production and thus the antifungal capacity of neutrophils and macrophages, and by decreasing the hyper-inflammation that leads to severe host damage. Leucocyte recruitment to infected kidneys and production of inflammatory mediators are decreased in p38γ/δ-null mice, reducing septic shock. p38γ/p38δ in myeloid cells are critical for this effect. Moreover, pharmacological inhibition of p38γ/p38δ in mice reduces fungal burden, revealing that these p38MAPKs may be therapeutic targets for treating C. albicans infection in humans.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Proteína Quinasa 12 Activada por Mitógenos/inmunología , Proteína Quinasa 13 Activada por Mitógenos/inmunología , Células Mieloides/inmunología , Animales , Candida albicans/fisiología , Candidiasis/genética , Candidiasis/microbiología , Femenino , Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 12 Activada por Mitógenos/deficiencia , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/deficiencia , Proteína Quinasa 13 Activada por Mitógenos/genética , Células Mieloides/metabolismo , Células Mieloides/microbiología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Óxido Nítrico Sintasa de Tipo II/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología
18.
Front Immunol ; 9: 65, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29434594

RESUMEN

p38 mitogen-activated protein kinase (MAPK) signal transduction pathways are essential regulators of the immune response. Particularly, p38γ and p38δ regulate many immune cell functions such as cytokine production, migration, or T cell activation; however, their involvement in immune cell development is largely unknown. Here, we analysed the role of p38 MAPK isoforms p38γ and p38δ in T cell differentiation in the thymus and in lymph nodes, using mice deficient in p38γ, p38δ, or in both. We found that the T cell differentiation program in the thymus was affected at different stages in p38γ-, p38δ-, and p38γ/δ-deficient mice, and also peripheral T cell homaeostasis was compromised. Particularly, p38δ deletion affects different stages of early CD4-CD8- double-negative thymocyte development, whereas lack of p38γ favours thymocyte positive selection from CD4+CD8+ double-positive to CD4+ or CD8+ single-positive cells. Our results identify unreported functions for p38γ and p38δ in T cells.


Asunto(s)
Diferenciación Celular , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Biomarcadores , Técnicas de Silenciamiento del Gen , Tejido Linfoide/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Timocitos/citología , Timocitos/metabolismo
19.
Science ; 354(6314): 904-908, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27856911

RESUMEN

Amyloid-ß (Aß) toxicity in Alzheimer's disease (AD) is considered to be mediated by phosphorylated tau protein. In contrast, we found that, at least in early disease, site-specific phosphorylation of tau inhibited Aß toxicity. This specific tau phosphorylation was mediated by the neuronal p38 mitogen-activated protein kinase p38γ and interfered with postsynaptic excitotoxic signaling complexes engaged by Aß. Accordingly, depletion of p38γ exacerbated neuronal circuit aberrations, cognitive deficits, and premature lethality in a mouse model of AD, whereas increasing the activity of p38γ abolished these deficits. Furthermore, mimicking site-specific tau phosphorylation alleviated Aß-induced neuronal death and offered protection from excitotoxicity. Our work provides insights into postsynaptic processes in AD pathogenesis and challenges a purely pathogenic role of tau phosphorylation in neuronal toxicity.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Neurotoxinas/antagonistas & inhibidores , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Animales , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Guanilato-Quinasas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Transducción de Señal
20.
J Biol Chem ; 291(32): 16699-708, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27246854

RESUMEN

The human protein tyrosine phosphatase non-receptor type 4 (PTPN4) prevents cell death induction in neuroblastoma and glioblastoma cell lines in a PDZ·PDZ binding motifs-dependent manner, but the cellular partners of PTPN4 involved in cell protection are unknown. Here, we described the mitogen-activated protein kinase p38γ as a cellular partner of PTPN4. The main contribution to the p38γ·PTPN4 complex formation is the tight interaction between the C terminus of p38γ and the PDZ domain of PTPN4. We solved the crystal structure of the PDZ domain of PTPN4 bound to the p38γ C terminus. We identified the molecular basis of recognition of the C-terminal sequence of p38γ that displays the highest affinity among all endogenous partners of PTPN4. We showed that the p38γ C terminus is also an efficient inducer of cell death after its intracellular delivery. In addition to recruiting the kinase, the binding of the C-terminal sequence of p38γ to PTPN4 abolishes the catalytic autoinhibition of PTPN4 and thus activates the phosphatase, which can efficiently dephosphorylate the activation loop of p38γ. We presume that the p38γ·PTPN4 interaction promotes cellular signaling, preventing cell death induction.


Asunto(s)
Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Complejos Multienzimáticos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 4/metabolismo , Transducción de Señal/fisiología , Muerte Celular , Línea Celular Tumoral , Humanos , Proteína Quinasa 12 Activada por Mitógenos/genética , Complejos Multienzimáticos/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 4/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA