Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 956
Filtrar
1.
Antiviral Res ; 226: 105891, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38649071

RESUMEN

Zoonoses such as ZIKV and SARS-CoV-2 pose a severe risk to global health. There is urgent need for broad antiviral strategies based on host-targets filling gaps between pathogen emergence and availability of therapeutic or preventive strategies. Significant reduction of pathogen titers decreases spread of infections and thereby ensures health systems not being overloaded and public life to continue. Based on previously observed interference with FGFR1/2-signaling dependent impact on interferon stimulated gene (ISG)-expression, we identified Pim kinases as promising druggable cellular target. We therefore focused on analyzing the potential of pan-Pim kinase inhibition to trigger a broad antiviral response. The pan-Pim kinase inhibitor AZD1208 exerted an extraordinarily high antiviral effect against various ZIKV isolates, SARS-CoV-2 and HBV. This was reflected by strong reduction in viral RNA, proteins and released infectious particles. Especially in case of SARS-CoV-2, AZD1208 led to a complete removal of viral traces in cells. Kinome-analysis revealed vast changes in kinase landscape upon AZD1208 treatment, especially for inflammation and the PI3K/Akt-pathway. For ZIKV, a clear correlation between antiviral effect and increase in ISG-expression was observed. Based on a cell culture model with impaired ISG-induction, activation of the PI3K-Akt-mTOR axis, leading to major changes in the endolysosomal equilibrium, was identified as second pillar of the antiviral effect triggered by AZD1208-dependent Pim kinase inhibition, also against HBV. We identified Pim-kinases as cellular target for a broad antiviral activity. The antiviral effect exerted by inhibition of Pim kinases is based on at least two pillars: innate immunity and modulation of the endolysosomal system.


Asunto(s)
Antivirales , Inmunidad Innata , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-pim-1 , SARS-CoV-2 , Transducción de Señal , Serina-Treonina Quinasas TOR , Humanos , Inmunidad Innata/efectos de los fármacos , Antivirales/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Virus Zika/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Animales , Virus de la Hepatitis B/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Línea Celular , COVID-19/inmunología , COVID-19/virología , Tratamiento Farmacológico de COVID-19 , Replicación Viral/efectos de los fármacos , Compuestos de Bifenilo , Tiazolidinas
2.
Neoplasia ; 52: 100996, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38593698

RESUMEN

Papillary thyroid carcinoma (PTC) is the most common endocrine malignancy, and its incidence has increased rapidly in recent years. The BRAF inhibitor vemurafenib is effective against BRAFV600E-positive PTC; however, acquired resistance to single agent therapy frequently leads to tumor recurrence and metastasis, underscoring the need to develop tailored treatment strategies. We previously showed that the oncogenic kinase PIM1 was associated with the malignant phenotype and prognosis of PTC. In this study, we showed that sustained expression of the PIM1 protein in PTC was affected by the BRAFV600E mutation. Based on this regulatory mechanism, we tested the synergistic effects of inhibitors of BRAF (BRAFi) and PIM1 in BRAFV600E-positive PTC cell lines and xenograft tumors. LC-MS metabolomics analyses suggested that BRAFi/PIMi therapy acted by restricting the amounts of critical amino acids and nucleotides required by cancer cells as well as modulating DNA methylation. This study elucidates the role of BRAFV600E in the regulation of PIM1 in PTC and demonstrates the synergistic effect of a novel combination, BRAFi/PIMi, for the treatment of PTC. This discovery, along with the pathways that may be involved in the powerful efficacy of BRAFi/PIMi strategy from the perspective of cell metabolism, provides insight into the molecular basis of PTC progression and offers new perspectives for BRAF-resistant PTC treatment.


Asunto(s)
Sinergismo Farmacológico , Mutación , Proteínas Proto-Oncogénicas B-raf , Proteínas Proto-Oncogénicas c-pim-1 , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Proteínas Proto-Oncogénicas c-pim-1/genética , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/tratamiento farmacológico , Cáncer Papilar Tiroideo/patología , Cáncer Papilar Tiroideo/metabolismo , Animales , Ratones , Línea Celular Tumoral , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Modelos Animales de Enfermedad , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico
3.
In Vitro Cell Dev Biol Anim ; 60(3): 278-286, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38485819

RESUMEN

Previous studies have implicated targeting Pim-1 proto-oncogene, serine/threonine kinase (PIM1) as a preventive measure against high glucose-induced cellular stress and apoptosis. This study aimed to reveal the potential role and regulatory mechanism of PIM1 in diabetic retinopathy. Human retinal microvascular endothelial cells (hRMECs) underwent high glucose induction, and fluctuations in PIM1 levels were assessed. By overexpressing PIM1, its effects on the levels of inflammatory factors, oxidative stress indicators, migration and tube formation abilities, tight junction protein expression levels, and ferroptosis in hRMECs were identified. Afterwards, hRMECs were treated with the ferroptosis-inducing agent erastin, and the effect of erastin on the above PIM1 regulatory functions was focused on. PIM1 was downregulated upon high glucose, and its overexpression inhibited the inflammatory response, oxidative stress, cell migration, and tube formation potential in hRMECs, whereas elevated tight junction protein levels. Furthermore, PIM1 overexpression reduced intracellular iron ion levels, lipid peroxidation, and levels of proteins actively involved in ferroptosis. Erastin treatment reversed the impacts of PIM1 on hRMECs, suggesting the mediation of ferroptosis in PIM1 regulation. The current study has yielded critical insights into the role of PIM1 in ameliorating high glucose-induced hRMEC dysfunction through the inhibition of ferroptosis.


Asunto(s)
Células Endoteliales , Ferroptosis , Humanos , Animales , Retina/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Glucosa/toxicidad , Glucosa/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/farmacología
4.
Int J Mol Sci ; 25(6)2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38542097

RESUMEN

The Proviral Integration site for the Moloney murine leukemia virus (PIM)-1 kinase and its family members (PIM-2 and PIM-3) regulate several cellular functions including survival, proliferation, and apoptosis. Recent studies showed their involvement in the pathogenesis of rheumatoid arthritis RA, while no studies are available on psoriatic arthritis (PsA) and axial spondyloarthritis (axSpA). The main objective of this study is to assess the expression of PIM kinases in inflammatory arthritides, their correlation with proinflammatory cytokines, and their variation after treatment with biologic disease-modifying anti-rheumatic drugs or JAK inhibitors. We evaluated PIM-1, -2, and -3 expression at the gene and protein level, respectively, in the peripheral blood mononuclear cells and serum of patients with RA, PsA, axSpA, and healthy individuals (CTR). All the samples showed expression of PIM-1, -2, and -3 kinases both at the gene and protein level. PIM-1 was the most expressed protein, PIM-3 the least. PIM kinase levels differed between controls and disease groups, with reduced PIM-1 protein and increased PIM-3 protein in all disease samples compared to controls. No difference was found in the expression of these molecules between the three different pathologies. PIM levels were not modified after 6 months of therapy. In conclusion, our preliminary data suggest a deregulation of the PIM pathway in inflammatory arthritides. In-depth studies on the role of PIM kinases in this field are warranted.


Asunto(s)
Artritis Psoriásica , Espondiloartritis Axial , Proteínas Proto-Oncogénicas c-pim-1 , Ratones , Animales , Humanos , Leucocitos Mononucleares , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Artritis Psoriásica/tratamiento farmacológico , Artritis Psoriásica/genética
5.
Medicine (Baltimore) ; 103(6): e36269, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38335426

RESUMEN

Sequence studies of the entire exome and transcriptome of lymphoma tissues have identified MYD88 and PIM1 as involved in the development and oncogenic signaling. We aimed to determine the frequency of MYD88 and PIM1 mutations, as well as their expressions in conjunction with the clinicopathological parameters identified in mature large B-cell non-Hodgkin lymphomas. The ten-year retrospective study included 50 cases of mature large B-cell lymphoma, diagnosed at the Pathology Department of the Emergency County Hospital of Constanta and Sacele County Hospital of Brasov. They were statistically analyzed by demographic, clinicopathological, and morphogenetic characteristics. We used a real-time polymerase chain reaction technique to identify PIM1 and MYD88 mutations as well as an immunohistochemical technique to evaluate the expressions of the 2 genes. Patients with lymphoma in the small bowel, spleen, brain, and testis had a low-performance status Eastern Cooperative Oncology Group (P = .001). The Eastern Cooperative Oncology Group performance status represented an independent risk factor predicting mortality (HR = 9.372, P < .001). An increased lactate dehydrogenase value was associated with a low survival (P = .002). The international prognostic index score represents a negative risk factor in terms of patient survival (HR = 4.654, P < .001). In cases of diffuse large B-cell lymphoma (DLBCL), immunopositivity of MYD88 is associated with non-germinal center B-cell origin (P < .001). The multivariate analysis observed the association between high lactate dehydrogenase value and the immunohistochemical expression of PIM1 or with the mutant status of the PIM1 gene representing negative prognostic factors (HR = 2.066, P = .042, respectively HR = 3.100, P = .004). In conclusion, our preliminary data suggest that the oncogenic mutations of PIM1 and MYD88 in our DLBCL cohort may improve the diagnosis and prognosis of DLBCL patients in an advanced stage.


Asunto(s)
Linfoma de Células B Grandes Difuso , Factor 88 de Diferenciación Mieloide , Masculino , Humanos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Estudios Retrospectivos , Pronóstico , Linfoma de Células B Grandes Difuso/patología , Lactato Deshidrogenasas/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/genética , Proteínas Proto-Oncogénicas c-pim-1/metabolismo
6.
Cell Immunol ; 397-398: 104805, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38244265

RESUMEN

PIM kinases are over-expressed by a number of solid malignancies including breast cancer, and are thought to regulate proliferation, survival, and resistance to treatment, making them attractive therapeutic targets. Because PIM kinases sit at the nexus of multiple oncodriver pathways, PIM antagonist drugs are being tested alone and in conjunction with other therapies to optimize outcomes. We therefore sought to test the combination of pharmacological PIM antagonism and Th1-associated immunotherapy. We show that the pan PIM antagonist, AZD1208, when combined in vitro with Th1 cytokines IFN-γ and TNF-α, potentiates metabolic suppression, overall cell death, and expression of apoptotic markers in human breast cancer cell lines of diverse phenotypes (HER-2pos/ERneg, HER-2pos/ERpos and triple-negative). Interestingly, AZD1208 was shown to moderately inhibit IFN-γ secretion by stimulated T lymphocytes of both human and murine origin, suggesting some inherent immunosuppressive activity of the drug. Nonetheless, when multiplexed therapies were tested in a murine model of HER-2pos breast cancer, combinations of HER-2 peptide-pulsed DCs and AZD1208, as well as recombinant IFN-γ plus AZD1208 significantly suppressed tumor outgrowth compared with single-treatment and control groups. These studies suggest that PIM antagonism may combine productively with certain immunotherapies to improve responsiveness.


Asunto(s)
Antineoplásicos , Compuestos de Bifenilo , Neoplasias , Proteínas Proto-Oncogénicas c-pim-1 , Tiazolidinas , Humanos , Animales , Ratones , Citocinas , Antineoplásicos/farmacología , Inmunoterapia , Proliferación Celular , Línea Celular Tumoral , Inhibidores de Proteínas Quinasas/farmacología
7.
J Med Chem ; 67(1): 38-64, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38164076

RESUMEN

Proviral integration sitea for Moloney-murine leukemia virus (PIM) kinases are a family of highly conserved serine/tyrosine kinases consisting of three members, PIM-1, PIM-2, and PIM-3. These kinases regulate a wide range of substrates through phosphorylation and affect key cellular processes such as transcription, translation, proliferation, apoptosis, and energy metabolism. Several PIM inhibitors are currently undergoing clinical trials, such as a phase I clinical trial of Uzanserti (5) for the treatment of relapsed diffuse large B-cell lymphoma that has been completed. The current focus encompasses the structural and biological characterization of PIM, ongoing research progress on small-molecule inhibitors undergoing clinical trials, and evaluation analysis of persisting challenges in this field. Additionally, the design and discovery of small-molecule inhibitors targeting PIM in recent years have been explored, with a particular emphasis on medicinal chemistry, aiming to provide valuable insights for the future development of PIM inhibitors.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas c-pim-1 , Ratones , Animales , Humanos , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fosforilación , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/química
8.
J Enzyme Inhib Med Chem ; 39(1): 2304044, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38230430

RESUMEN

New aromatic O-alkyl pyridine derivatives were designed and synthesised as Proviral Integration Moloney (PIM)-1 kinase inhibitors. 4c and 4f showed potent in vitro anticancer activity against NFS-60, HepG-2, PC-3, and Caco-2 cell lines and low toxicity against normal human lung fibroblast Wi-38 cell line. Moreover, 4c and 4f induced apoptosis in the four tested cancer cell lines with high percentage. In addition, 4c and 4f significantly induced caspase 3/7 activation in HepG-2 cell line. Furthermore, 4c and 4f showed potent PIM-1 kinase inhibitory activity with IC50 = 0.110, 0.095 µM, respectively. Kinetic studies indicated that 4c and 4f were both competitive and non-competitive inhibitors for PIM-1 kinase enzyme. In addition, in silico prediction of physiochemical properties, pharmacokinetic profile, ligand efficiency, ligand lipophilic efficiency, and induced fit docking studies were consistent with the biological and kinetic studies, and predicted that 4c and 4f could act as PIM-1 kinase competitive non-adenosine triphosphate (ATP) mimetics with drug like properties.


Asunto(s)
Antineoplásicos , Piridonas , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Proteínas Proto-Oncogénicas c-pim-1/química , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Inhibidores de Proteínas Quinasas/química , Células CACO-2 , Cinética , Ligandos , Apoptosis , Proliferación Celular , Simulación del Acoplamiento Molecular , Ensayos de Selección de Medicamentos Antitumorales , Relación Estructura-Actividad
9.
J Biomol Struct Dyn ; 42(5): 2392-2409, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37160699

RESUMEN

Cancer is an unimpeded growth of cells leading to metathesis of cancer and eventually spread throughout the body. PIM kinases are the members of the serine threonine kinase playing role in cancer progression, differentiation and proliferation. Till date there is no single drug targeting PIM-1 kinase in the market, that has made itself a target in limelight for the discover of new anticancer agents. The contemporary research focusses on the development of new inhibitors of PIM-1 kinase by application of ligand-based and structure-based perspective of drug discovery namely 3D-QSAR, molecular docking and dynamics. The following study stated the correlation amid structural and biological activity of the compounds employing 3D-QSAR analysis. Three 3D-QSAR models were generated using 33 molecules from which the excellent model stated an encouraging conventional correlation coefficient (r2) 0.8651, cross validation coefficient (q2) 0.7609. Furthermore, the predicted correlation coefficient (r2 pred) 0.6274, respectively. Molecular docking studies revealed that the most active compound 26 resided in the active pocket of PIM-1 kinase establishing hydrogen bond interactions with Asp186 in the DFG motif; similarly, all other molecules were engaged within the active site of the PIM-1 kinase. Moreover, molecular dynamics simulation study stated the stability of the ligand in the active site of PIM-1 kinase protein by developing two hydrogen bonds throughout the trajectory of 100 ns. In nutshell, the output stated the successful application of ligand and structure-based strategy for the development of novel PIM-1 kinase inhibitors as anticancer agents.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Simulación de Dinámica Molecular , Simulación del Acoplamiento Molecular , Proteínas Proto-Oncogénicas c-pim-1 , Relación Estructura-Actividad Cuantitativa , Ligandos , Antineoplásicos/farmacología
10.
Eur J Med Chem ; 264: 116016, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38071792

RESUMEN

PIM kinases, a serine/threonine kinase family with three isoforms, has been well-known to participate in multiple physiological processes by phosphorylating various downstream targets. Accumulating evidence has recently unveiled that aberrant upregulation of PIM kinases (PIM1, PIM2, and PIM3) are closely associated with tumor cell proliferation, migration, survival, and even resistance. Inhibiting or silencing of PIM kinases has been reported have remarkable antitumor effects, such as anti-proliferation, pro-apoptosis and resensitivity, indicating the therapeutic potential of PIM kinases as potential druggable targets in many types of human cancers. More recently, several pharmacological small-molecule inhibitors have been preclinically and clinically evaluated and showed their therapeutic potential; however, none of them has been approved for clinical application so far. Thus, in this perspective, we focus on summarizing the oncogenic roles of PIM kinases, key signaling network, and pharmacological small-molecule inhibitors, which will provide a new clue on discovering more candidate antitumor drugs targeting PIM kinases in the future.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas c-pim-1 , Humanos , Proteínas Serina-Treonina Quinasas , Neoplasias/tratamiento farmacológico , Transducción de Señal , Proliferación Celular , Inhibidores de Proteínas Quinasas/farmacología
11.
Naunyn Schmiedebergs Arch Pharmacol ; 397(2): 1219-1231, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37658212

RESUMEN

Smooth muscle contraction by Pim kinases and ZIPK has been suggested, but evidence for lower urinary tract organs or using Pim-selective inhibitor concentrations is not yet available. Here, we assessed effects of the Pim inhibitors AZD1208 and TCS PIM-1 and the dual ZIPK/Pim inhibitor HS38 on contractions of human prostate and bladder tissues and of porcine interlobar arteries. Human tissues were obtained from radical prostatectomy and radical cystectomy and renal interlobar arteries from pigs. Contractions were studied in an organ bath. Noradrenaline-, phenylephrine- and methoxamine-induced contractions were reduced (up to > 50%) with 500-nM AZD1208 in prostate tissues and to lesser degree and not consistently with all agonists in interlobar arteries. A total of 100-nM AZD1208 or 500-nM TCS PIM-1 did not affect agonist-induced contractions in prostate tissues. Decreases in agonist-induced contractions with 3-µM HS38 in prostate tissues and interlobar arteries were of small extent and did not occur with each agonist. Carbachol-induced contractions in detrusor tissues were unchanged with AZD1208 (500 nM) or HS38. Electric field stimulation-induced contractions were not affected with AZD1208 or HS38 in any tissue, but slightly reduced with 500-nM TCS PIM-1 in prostate tissues. Concentration-dependent effects of Pim inhibitors suggest lacking Pim-driven smooth muscle contraction in the prostate, bladder, and interlobar arteries but point to organ-specific functions of off-targets. Procontractile functions of ZIPK in the prostate and interlobar arteries may be limited and are lacking in the detrusor.


Asunto(s)
Compuestos de Bifenilo , Músculo Liso Vascular , Próstata , Proteínas Proto-Oncogénicas c-pim-1 , Tiazolidinas , Masculino , Humanos , Animales , Porcinos , Vejiga Urinaria , Proteínas Quinasas Asociadas a Muerte Celular/farmacología , Contracción Muscular
12.
Cell Chem Biol ; 31(2): 326-337.e11, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38016478

RESUMEN

PIM kinases have important pro-tumorigenic roles and mediate several oncogenic traits, including cell proliferation, survival, and chemotherapeutic resistance. As a result, multiple PIM inhibitors have been pursued as investigational new drugs in cancer; however, response to PIM inhibitors in solid tumors has fallen short of expectations. We found that inhibition of PIM kinase activity stabilizes protein levels of all three PIM isoforms (PIM1/2/3), and this can promote resistance to PIM inhibitors and chemotherapy. To overcome this effect, we designed PIM proteolysis targeting chimeras (PROTACs) to target PIM for degradation. PIM PROTACs effectively downmodulated PIM levels through the ubiquitin-proteasome pathway. Importantly, degradation of PIM kinases was more potent than inhibition of catalytic activity at inducing apoptosis in prostate cancer cell line models. In conclusion, we provide evidence of the advantages of degrading PIM kinases versus inhibiting their catalytic activity to target the oncogenic functions of PIM kinases.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias de la Próstata , Masculino , Humanos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Fosforilación , Apoptosis , Proliferación Celular , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-pim-1
13.
Int Immunopharmacol ; 126: 111256, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37992447

RESUMEN

The inflammatory response runs through the whole pathogenesis of systemic lupus erythematosus (SLE). Mesenchymal stem cells (MSC) have exhibited a positive therapeutic effect on SLE. This study aimed to ascertain the pathogenic role of inflammasome activation in SLE and whether MSC alleviate SLE by suppressing it. The results showed that the nucleotide-binding oligomerization domain-like receptor 3 (NLRP3) inflammasome was activated in macrophages from MRL/lpr mice and patients with SLE, correlating with disease activity. After MSC transplantation, the disease severity in MRL/lpr mice was alleviated, and NLRP3 inflammasome activation was inhibited with decreased levels of NLRP3 and caspase-1 in macrophages. Furthermore, lower serum levels of interleukin (IL)-1ß and IL-18 were observed in patients with SLE who underwent MSC transplantation. In vitro and in vivo studies indicated that MSC suppressed NLRP3 inflammasome activation by inhibiting Pim-1 expression. The findings provide an updated view of inflammasome signaling in SLE. Additionally, MSC ameliorated SLE by inhibiting NLRP3 inflammasome activation, implying a possible molecular mechanism for the clinical application of MSC and a potential therapeutic target in patients with SLE.


Asunto(s)
Lupus Eritematoso Sistémico , Células Madre Mesenquimatosas , Animales , Humanos , Ratones , Inflamasomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos MRL lpr , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas Proto-Oncogénicas c-pim-1
14.
Oncogene ; 43(6): 406-419, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38097734

RESUMEN

Lipid droplets (LDs) are dynamic organelles with a neutral lipid core surrounded by a phospholipid monolayer. Solid tumors exhibit LD accumulation, and it is believed that LDs promote cell survival by providing an energy source during energy deprivation. However, the precise mechanisms controlling LD accumulation and utilization in prostate cancer are not well known. Here, we show peroxisome proliferator-activated receptor α (PPARα) acts downstream of PIM1 kinase to accelerate LD accumulation and promote cell proliferation in prostate cancer. Mechanistically, PIM1 inactivates glycogen synthase kinase 3 beta (GSK3ß) via serine 9 phosphorylation. GSK3ß inhibition stabilizes PPARα and enhances the transcription of genes linked to peroxisomal biogenesis (PEX3 and PEX5) and LD growth (Tip47). The effects of PIM1 on LD accumulation are abrogated with GW6471, a specific inhibitor for PPARα. Notably, LD accumulation downstream of PIM1 provides a significant survival advantage for prostate cancer cells during nutrient stress, such as glucose depletion. Inhibiting PIM reduces LD accumulation in vivo alongside slow tumor growth and proliferation. Furthermore, TKO mice, lacking PIM isoforms, exhibit suppression in circulating triglycerides. Overall, our findings establish PIM1 as an important regulator of LD accumulation through GSK3ß-PPARα signaling axis to promote cell proliferation and survival during nutrient stress.


Asunto(s)
Gotas Lipídicas , Neoplasias de la Próstata , Masculino , Humanos , Animales , Ratones , Glucógeno Sintasa Quinasa 3 beta , Gotas Lipídicas/patología , PPAR alfa/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proliferación Celular , Proteínas Proto-Oncogénicas c-pim-1/genética
15.
Cell Rep ; 42(12): 113469, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-38039135

RESUMEN

The serine/threonine-specific Moloney murine leukemia virus (PIM) kinase family (i.e., PIM1, PIM2, and PIM3) has been extensively studied in tumorigenesis. PIM kinases are downstream of several cytokine signaling pathways that drive immune-mediated diseases. Uncontrolled T helper 17 (Th17) cell activation has been associated with the pathogenesis of autoimmunity. However, the detailed molecular function of PIMs in human Th17 cell regulation has yet to be studied. In the present study, we comprehensively investigated how the three PIMs simultaneously alter transcriptional gene regulation during early human Th17 cell differentiation. By combining PIM triple knockdown with bulk and scRNA-seq approaches, we found that PIM deficiency promotes the early expression of key Th17-related genes while suppressing Th1-lineage genes. Further, PIMs modulate Th cell signaling, potentially via STAT1 and STAT3. Overall, our study highlights the inhibitory role of PIMs in human Th17 cell differentiation, thereby suggesting their association with autoimmune phenotypes.


Asunto(s)
Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-pim-1 , Animales , Ratones , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/genética , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Transducción de Señal , Hematopoyesis , Diferenciación Celular , Células Th17/metabolismo
16.
Cell Mol Biol (Noisy-le-grand) ; 69(9): 183-188, 2023 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-37807309

RESUMEN

Lower extremity deep venous thrombosis (LEDVT) has a high incidence and mortality. Crocin has the potential to ameliorate thrombosis. The study aimed to clarify whether crocin affects LEDVT. Human umbilical vein endothelial cells (HUVECs) were exposed to thrombin and crocin (0, 5, 10, 20, 40, and 80 µM). Cell viability was assessed by MTT assay. Cellular behaviors were assessed using flow cytometry, TUNEL assay, and tube formation assay. The binding relationship between crocin and PIM1 was analyzed by molecular docking. The underlying mechanism of PIM1 was determined by reverse transcription-quantitative PCR, dual-luciferase reporter assay, and RIP. We found that crocin (5, 10, 20, and 40 µM) promoted thrombin-treated HUVEC viability in a dose-dependent manner. Crocin inhibited apoptosis and promoted the angiogenesis of HUVECs induced by thrombin. PIM1 was a target of crocin and was upregulated in patients with LEDVT and thrombin-treated cells. Foxo3a could interact with PIM1 and positively related to PIM1 expression. Moreover, the knockdown of PIM1 suppressed apoptosis and promoted angiogenesis in thrombin-HUVECs treated with crocin, while overexpression of Foxo3a reversed the effects. In conclusion, crocin inhibited apoptosis and promoted the angiogenesis of HUVECs induced by thrombin via the PIM1/Foxo3a axis, suggesting that crocin may be effective for LEDVT therapy.


Asunto(s)
MicroARNs , Trombosis de la Vena , Humanos , Simulación del Acoplamiento Molecular , Trombina/metabolismo , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Extremidad Inferior/irrigación sanguínea , MicroARNs/metabolismo , Apoptosis , Proteínas Proto-Oncogénicas c-pim-1/metabolismo
17.
Science ; 382(6666): eadg2253, 2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37797010

RESUMEN

Disruption of cellular activities by pathogen virulence factors can trigger innate immune responses. Interferon-γ (IFN-γ)-inducible antimicrobial factors, such as the guanylate binding proteins (GBPs), promote cell-intrinsic defense by attacking intracellular pathogens and by inducing programmed cell death. Working in human macrophages, we discovered that GBP1 expression in the absence of IFN-γ killed the cells and induced Golgi fragmentation. IFN-γ exposure improved macrophage survival through the activity of the kinase PIM1. PIM1 phosphorylated GBP1, leading to its sequestration by 14-3-3σ, which thereby prevented GBP1 membrane association. During Toxoplasma gondii infection, the virulence protein TgIST interfered with IFN-γ signaling and depleted PIM1, thereby increasing GBP1 activity. Although infected cells can restrain pathogens in a GBP1-dependent manner, this mechanism can protect uninfected bystander cells. Thus, PIM1 can provide a bait for pathogen virulence factors, guarding the integrity of IFN-γ signaling.


Asunto(s)
Proteínas de Unión al GTP , Interacciones Huésped-Patógeno , Inmunidad Innata , Interferón gamma , Proteínas Proto-Oncogénicas c-pim-1 , Toxoplasma , Toxoplasmosis , Humanos , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Interferón gamma/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Toxoplasmosis/inmunología , Factores de Virulencia/metabolismo , Macrófagos/inmunología , Proteínas 14-3-3/metabolismo , Interacciones Huésped-Patógeno/inmunología
18.
Cancer Biol Ther ; 24(1): 2246208, 2023 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-37621144

RESUMEN

Significant improvement in targeted therapy for colorectal cancer (CRC) has occurred over the past few decades since the approval of the EGFR inhibitor cetuximab. However, cetuximab is used only for patients possessing the wild-type oncogene KRAS, NRAS, and BRAF, and even most of these eventually acquire therapeutic resistance, via activation of parallel oncogenic pathways such as RAS-MAPK or PI3K/Akt/mTOR. The two aforementioned pathways also contribute to the development of therapeutic resistance in CRC patients, due to compensatory and feedback mechanisms. Therefore, combination drug therapies (versus monotherapy) targeting these multiple pathways may be necessary for further efficacy against CRC. In this study, we identified PIK3CA mutant (PIK3CA MT) as a determinant of resistance to SMI-4a, a highly selective PIM1 kinase inhibitor, in CRC cell lines. In CRC cell lines, SMI-4a showed its effect only in PIK3CA wild type (PIK3CA WT) cell lines, while PIK3CA MT cells did not respond to SMI-4a in cell death assays. In vivo xenograft and PDX experiments confirmed that PIK3CA MT is responsible for the resistance to SMI-4a. Inhibition of PIK3CA MT by PI3K inhibitors restored SMI-4a sensitivity in PIK3CA MT CRC cell lines. Taken together, these results demonstrate that sensitivity to SMI-4a is determined by the PIK3CA genotype and that co-targeting of PI3K and PIM1 in PIK3CA MT CRC patients could be a promising and novel therapeutic approach for refractory CRC patients.


Asunto(s)
Neoplasias del Colon , Fosfatidilinositol 3-Quinasas , Humanos , Cetuximab/farmacología , Cetuximab/uso terapéutico , Fosfatidilinositol 3-Quinasas/genética , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Biomarcadores , Fosfatidilinositol 3-Quinasa Clase I/genética , Proteínas Proto-Oncogénicas c-pim-1/genética
19.
Anal Chem ; 95(31): 11732-11740, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37490364

RESUMEN

Currently, kinase inhibitors have been applied in the diagnosis or treatment of cancer with their unique advantages. It is of great significance to develop some comprehensive theranostic reagents based on kinase inhibitors to improve the performance of reagents for biomedical applications. Besides, tracking changes in the intracellular environment (e.g., pH) during cancer development and drug delivery is also critical for cancer research and treatment. Therefore, it is an urgent desire to design some novel multifunctional reagents based on kinase inhibitor strategies that can trace changes in the microenvironment of cancer cells. In this paper, a multifunctional theranostic reagent based on Pim-1 kinase inhibitor 5-bromobenzofuran-2-carboxylic acid is proposed. The theranostic probe binds to tumor-specific Pim-1 kinase, releases strong fluorescence, and produces cytotoxicity, thus achieving cell screening and killing effects. Furthermore, the probe can specifically target lysosomes and sensitively respond to pH. It can be used to track the pH changes in the intracellular environment under conditions of autophagy and external stimulation, as a visual tool to monitor pH fluctuations during cancer treatment. In conclusion, this simple but multifunctional theranostic reagent proposed in this work is expected to provide a promising method for cancer diagnosis and therapy.


Asunto(s)
Antineoplásicos , Proteínas Proto-Oncogénicas c-pim-1 , Medicina de Precisión , Línea Celular Tumoral , Antineoplásicos/farmacología , Nanomedicina Teranóstica/métodos , Concentración de Iones de Hidrógeno
20.
Int J Mol Sci ; 24(14)2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37511341

RESUMEN

Pim Kinases; Pim-1, Pim-2, and Pim-3, are a family of constitutively active serine/threonine kinases, widely associated with cell survival, proliferation, and migration. Historically considered to be functionally redundant, independent roles for the individual isoforms have been described. Whilst most established for their role in cancer progression, there is increasing evidence for wider pathological roles of Pim kinases within the context of cardiovascular disease, including inflammation, thrombosis, and cardiac injury. The Pim kinase isoforms have widespread expression in cardiovascular tissues, including the heart, coronary artery, aorta, and blood, and have been demonstrated to be upregulated in several co-morbidities/risk factors for cardiovascular disease. Pim kinase inhibition may thus be a desirable therapeutic for a multi-targeted approach to treat cardiovascular disease and some of the associated risk factors. In this review, we discuss what is known about Pim kinase expression and activity in cells of the cardiovascular system, identify areas where the role of Pim kinase has yet to be fully explored and characterised and review the suitability of targeting Pim kinase for the prevention and treatment of cardiovascular events in high-risk individuals.


Asunto(s)
Enfermedades Cardiovasculares , Humanos , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-pim-1 , Proteínas Serina-Treonina Quinasas/metabolismo , Isoformas de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA