Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
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.
Cells ; 12(13)2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37443708

RESUMEN

p38γ MAPK (also called ERK6 or SAPK3) is a family member of stress-activated MAPKs and has common and specific roles as compared to other p38 proteins in signal transduction. Recent studies showed that, in addition to inflammation, p38γ metabolic signaling is involved in physiological exercise and in pathogenesis of cancer, diabetes, and Alzheimer's disease, indicating its potential as a therapeutic target. p38γphosphorylates at least 19 substrates through which p38γ activity is further modified to regulate life-important cellular processes such as proliferation, differentiation, cell death, and transformation, thereby impacting biological outcomes of p38γ-driven pathogenesis. P38γ signaling is characterized by its unique reciprocal regulation with its specific phosphatase PTPH1 and by its direct binding to promoter DNAs, leading to transcriptional activation of targets including cancer-like stem cell drivers. This paper will review recent findings about p38γ inflammation and metabolic signaling in physiology and diseases. Moreover, we will discuss the progress in the development of p38γ-specific pharmacological inhibitors for therapeutic intervention in disease prevention and treatment by targeting the p38γ signaling network.


Asunto(s)
Proteína Quinasa 12 Activada por Mitógenos , Transducción de Señal , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Fosforilación
4.
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
5.
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
6.
Int J Mol Sci ; 24(8)2023 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-37108523

RESUMEN

Protein kinase p38γ is an attractive target against cancer because it plays a pivotal role in cancer cell proliferation by phosphorylating the retinoblastoma tumour suppressor protein. Therefore, inhibition of p38γ with active small molecules represents an attractive alternative for developing anti-cancer drugs. In this work, we present a rigorous and systematic virtual screening framework to identify potential p38γ inhibitors against cancer. We combined the use of machine learning-based quantitative structure activity relationship modelling with conventional computer-aided drug discovery techniques, namely molecular docking and ligand-based methods, to identify potential p38γ inhibitors. The hit compounds were filtered using negative design techniques and then assessed for their binding stability with p38γ through molecular dynamics simulations. To this end, we identified a promising compound that inhibits p38γ activity at nanomolar concentrations and hepatocellular carcinoma cell growth in vitro in the low micromolar range. This hit compound could serve as a potential scaffold for further development of a potent p38γ inhibitor against cancer.


Asunto(s)
Antineoplásicos , Simulación de Dinámica Molecular , Antineoplásicos/farmacología , Bioensayo , Descubrimiento de Drogas , Ligandos , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad Cuantitativa , Proteína Quinasa 12 Activada por Mitógenos/metabolismo
7.
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
8.
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
9.
Cell Death Dis ; 13(3): 210, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35246508

RESUMEN

It is urgent to identify and validate biomarkers for early diagnosis and efficient treatment of nasopharyngeal carcinoma (NPC). Recent studies have proposed p38 gamma (p38γ) as a cyclin-dependent kinase (CDK)-like kinase that phosphorylates retinoblastoma (Rb) to promote cyclins expression and tumorigenesis. Here the Gene Expression Profiling Interactive Analysis (GEPIA) database and results from the local NPC tissues demonstrate that p38γ is significantly upregulated in NPC tissues, correlating with poor overall survival. Furthermore, p38γ mRNA and protein expression is elevated in established NPC cell lines (CNE-1 HONE-1 and CNE-2) and primary human NPC cells, but low expression detected in human nasal epithelial cells. In established and primary NPC cells, p38γ depletion, using the shRNA strategy or the CRISPR/Cas9 gene-editing method, largely inhibited cell growth, proliferation and migration, and induced significant apoptosis activation. Contrarily, ectopic p38γ overexpression exerted opposite activity and promoted NPC cell proliferation and migration. Retinoblastoma (Rb) phosphorylation and cyclin E1/A expression were decreased in NPC cells with p38γ silencing or knockout, but increased after p38γ overexpression. Moreover, mitochondrial subcellular p38γ localization was detected in NPC cells. Significantly, p38γ depletion disrupted mitochondrial functions, causing mitochondrial depolarization, reactive oxygen species production, oxidative injury and ATP depletion in NPC cells. In vivo, intratumoral injection of adeno-associated virus-packed p38γ shRNA potently inhibited primary human NPC xenograft growth in nude mice. In p38γ shRNA virus-injected NPC xenograft tissues, p38γ expression, Rb phosphorylation, cyclin E1/A expression and ATP levels were dramatically decreased. Taken together, we conclude that p38γ overexpression is required for NPC cell growth, acting as a promising therapeutic target of NPC.


Asunto(s)
Neoplasias Nasofaríngeas , Neoplasias de la Retina , Retinoblastoma , Adenosina Trifosfato , Animales , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular/genética , Ciclinas , Humanos , Ratones , Ratones Desnudos , Proteína Quinasa 12 Activada por Mitógenos , Carcinoma Nasofaríngeo/tratamiento farmacológico , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/patología , ARN Interferente Pequeño/uso terapéutico
10.
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
11.
PLoS Biol ; 19(11): e3001447, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34758018

RESUMEN

During the first weeks of postnatal heart development, cardiomyocytes undergo a major adaptive metabolic shift from glycolytic energy production to fatty acid oxidation. This metabolic change is contemporaneous to the up-regulation and activation of the p38γ and p38δ stress-activated protein kinases in the heart. We demonstrate that p38γ/δ contribute to the early postnatal cardiac metabolic switch through inhibitory phosphorylation of glycogen synthase 1 (GYS1) and glycogen metabolism inactivation. Premature induction of p38γ/δ activation in cardiomyocytes of newborn mice results in an early GYS1 phosphorylation and inhibition of cardiac glycogen production, triggering an early metabolic shift that induces a deficit in cardiomyocyte fuel supply, leading to whole-body metabolic deregulation and maladaptive cardiac pathogenesis. Notably, the adverse effects of forced premature cardiac p38γ/δ activation in neonate mice are prevented by maternal diet supplementation of fatty acids during pregnancy and lactation. These results suggest that diet interventions have a potential for treating human cardiac genetic diseases that affect heart metabolism.


Asunto(s)
Glucógeno Sintasa/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Miocardio/enzimología , Animales , Animales Recién Nacidos , Cardiomegalia/enzimología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Dieta Alta en Grasa , Activación Enzimática , Conducta Alimentaria , Femenino , Eliminación de Gen , Intolerancia a la Glucosa/enzimología , Glucógeno/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Miocitos Cardíacos/enzimología , Especificidad de Órganos , Fosforilación
12.
FEBS Lett ; 595(20): 2570-2592, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34455585

RESUMEN

We describe here for the first time a lipid-binding-domain (LBD) in p38γ mitogen-activated protein kinase (MAPK) involved in the response of T cells to a newly identified inhibitor, CSH71. We describe how CSH71, which binds to both the LBD and the ATP-binding pocket of p38γ, is selectively cytotoxic to CTCL Hut78 cells but spares normal healthy peripheral blood mononuclear (PBMC) cells, and propose possible molecular mechanisms for its action. p38γ is a key player in CTCL development, and we expect that the ability to regulate its expression by specifically targeting the lipid-binding domain will have important clinical relevance. Our findings characterize novel mechanisms of gene regulation in T lymphoma cells and validate the use of computational screening techniques to identify inhibitors for therapeutic development.


Asunto(s)
Adenosina Trifosfato/metabolismo , Linfoma Cutáneo de Células T/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Neoplasias Cutáneas/metabolismo , Antineoplásicos/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma Cutáneo de Células T/tratamiento farmacológico , Linfoma Cutáneo de Células T/genética , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética
14.
Cell Metab ; 32(4): 643-653.e4, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32783890

RESUMEN

Metformin is the first-line therapy for type 2 diabetes, but there are large inter-individual variations in responses to this drug. Its mechanism of action is not fully understood, but activation of AMP-activated protein kinase (AMPK) and changes in the gut microbiota appear to be important. The inhibitory role of microbial metabolites on metformin action has not previously been investigated. Here, we show that concentrations of the microbial metabolite imidazole propionate are higher in subjects with type 2 diabetes taking metformin who have high blood glucose. We also show that metformin-induced glucose lowering is not observed in mice pretreated with imidazole propionate. Furthermore, we demonstrate that imidazole propionate inhibits AMPK activity by inducing inhibitory AMPK phosphorylation, which is dependent on imidazole propionate-induced basal Akt activation. Finally, we identify imidazole propionate-activated p38γ as a novel kinase for Akt and demonstrate that p38γ kinase activity mediates the inhibitory action of imidazole propionate on metformin.


Asunto(s)
Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Imidazoles/farmacología , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Línea Celular , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipoglucemiantes/farmacología , Imidazoles/administración & dosificación , Imidazoles/metabolismo , Inyecciones Intraperitoneales , Masculino , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos
15.
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
16.
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
17.
Biochemistry ; 58(51): 5160-5172, 2019 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-31794659

RESUMEN

The inactive state of mitogen-activated protein kinases (MAPKs) adopts an open conformation while the active state exists in a compact form stabilized by phosphorylation. In the active state, eukaryotic kinases undergo breathing motions related to substrate binding and product release that have not previously been detected in the inactive state. However, docking interactions of partner proteins with inactive MAPK kinases exhibit allostery in binding of activating kinases. Interactions at a site distant from the activation loop are coupled to the configuration of the activation loop, suggesting that the inactive state may also undergo concerted dynamics. X-ray crystallographic studies of nonphosphorylated, inactive p38γ reveal differences in domain orientations and active site structure in the two molecules in the asymmetric unit. One molecule resembles an inactive kinase with an open active site. The second molecule has a rotation of the N-lobe that leads to partial compaction of the active site, resulting in a conformation that is intermediate between the inactive open state and the fully closed state of the activated kinase. Although the compact state of apo p38γ displays several of the features of the activated enzyme, it remains catalytically inert. In solution, the kinase fluctuates on a millisecond time scale between the open ground state and a weakly populated excited state that is similar in structure to the compact state observed in the crystal. The nuclear magnetic resonance and crystal structure data imply that interconversion between the open and compact states involves a molecular switch associated with the DFG loop.


Asunto(s)
Proteína Quinasa 12 Activada por Mitógenos/química , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia Conservada , Activación Enzimática , Humanos , Modelos Moleculares , Dominios Proteicos
18.
Cells ; 8(11)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31683954

RESUMEN

Transient receptor potential canonical channel-6 (TRPC6) is one of the Ca2+-permeable non-selective cation channels. TRPC6 is mainly expressed in dentate granule cell (DGC), which is one of the most resistant neuronal populations to various harmful stresses. Although TRPC6 knockdown evokes the massive DGC degeneration induced by status epilepticus (a prolonged seizure activity, SE), the molecular mechanisms underlying the role of TRPC6 in DGC viability in response to SE are still unclear. In the present study, hyperforin (a TRPC6 activator) facilitated mitochondrial fission in DGC concomitant with increases in Lon protease-1 (LONP1, a mitochondrial protease) expression and extracellular-signal-regulated kinase 1/2 (ERK1/2) phosphorylation under physiological conditions, which were abrogated by U0126 (an ERK1/2 inhibitor) co-treatment. TRPC6 knockdown showed the opposite effects on LONP1 expression, ERK1/2 activity, and mitochondrial dynamics. In addition, TRPC6 siRNA and U0126 evoked the massive DGC degeneration accompanied by mitochondrial elongation following SE, independent of seizure severity. However, LONP1 siRNA exacerbated SE-induced DGC death without affecting mitochondrial length. These findings indicate that TRPC6-ERK1/2 activation may increase DGC invulnerability to SE by regulating LONP1 expression as well as mitochondrial dynamics. Therefore, TRPC6-ERK1/2-LONP1 signaling pathway will be an interesting and important therapeutic target for neuroprotection from various neurological diseases.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Proteasa La/metabolismo , Estado Epiléptico/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Butadienos/farmacología , Muerte Celular/efectos de los fármacos , Giro Dentado/metabolismo , Masculino , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/metabolismo , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/enzimología
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA