Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Int J Oral Sci ; 16(1): 20, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38418808

RESUMEN

Periodontitis is a common chronic inflammatory disease that causes the periodontal bone destruction and may ultimately result in tooth loss. With the progression of periodontitis, the osteoimmunology microenvironment in periodontitis is damaged and leads to the formation of pathological alveolar bone resorption. CD301b+ macrophages are specific to the osteoimmunology microenvironment, and are emerging as vital booster for conducting bone regeneration. However, the key upstream targets of CD301b+ macrophages and their potential mechanism in periodontitis remain elusive. In this study, we concentrated on the role of Tim4, a latent upstream regulator of CD301b+ macrophages. We first demonstrated that the transcription level of Timd4 (gene name of Tim4) in CD301b+ macrophages was significantly upregulated compared to CD301b- macrophages via high-throughput RNA sequencing. Moreover, several Tim4-related functions such as apoptotic cell clearance, phagocytosis and engulfment were positively regulated by CD301b+ macrophages. The single-cell RNA sequencing analysis subsequently discovered that Cd301b and Timd4 were specifically co-expressed in macrophages. The following flow cytometric analysis indicated that Tim4 positive expression rates in total macrophages shared highly synchronized dynamic changes with the proportions of CD301b+ macrophages as periodontitis progressed. Furthermore, the deficiency of Tim4 in mice decreased CD301b+ macrophages and eventually magnified alveolar bone resorption in periodontitis. Additionally, Tim4 controlled the p38 MAPK signaling pathway to ultimately mediate CD301b+ macrophages phenotype. In a word, Tim4 might regulate CD301b+ macrophages through p38 MAPK signaling pathway in periodontitis, which provided new insights into periodontitis immunoregulation as well as help to develop innovative therapeutic targets and treatment strategies for periodontitis.


Asunto(s)
Pérdida de Hueso Alveolar , Periodontitis , Animales , Ratones , Pérdida de Hueso Alveolar/metabolismo , Eferocitosis , Macrófagos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico , Periodontitis/tratamiento farmacológico
2.
Clin Respir J ; 18(1): e13729, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38286741

RESUMEN

BACKGROUND: Asthma associated with obesity is a chronic disease characterized by earlier airway remodeling, severe wheezing, and increased insensitivity to hormone therapy. Reticuline, a bioactive compound of Magnoliae Flos, exerts anti-inflammatory activity and can inhibit neutrophil recruitment. Thus, this study investigated the role of reticuline in obesity-related asthma. METHODS: The BALB/c mice fed a low-fat diet (LFD) and high-fat diet (HFD) were intranasally challenged with house dust mites (HDMs) or ovalbumin (OVA). Reticuline (0.25 mg/kg) was administrated into mice by intragastrical gavage. Airway hyper-responsiveness was examined after the final challenge. Body weight was measured, and bronchoalveolar lavage fluid (BALF) and lung tissues were collected. The number of inflammatory cells in BALF was estimated. Histological changes were assessed by performing hematoxylin-eosin staining, and production of proinflammatory cytokines and IgE was examined by ELISA kits. Related pathways were studied with western blotting. RESULTS: Reticuline suppressed airway resistance and inflammatory infiltration in lung tissue and reduced inflammatory cell recruitment in BALF in obesity mice with asthma. Additionally, the levels of IL-17A, IL-1ß, IL-5, macrophage inflammatory protein 2, and regulated on activation, normal T cell expressed and secreted in the lung were reduced by reticuline. Mechanistically, reticuline inactivated the JAK2/STAT3/SOCS3 and p38 MAPK/NF-κB signaling pathways in obesity-related asthma. CONCLUSION: Reticuline alleviates airway inflammation in obesity-related asthma by inactivating the JAK2/STAT3/SOCS3 and p38 MAPK/NF-κB signaling pathways.


Asunto(s)
Asma , Bencilisoquinolinas , Janus Quinasa 2 , FN-kappa B , Factor de Transcripción STAT3 , Animales , Ratones , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Asma/metabolismo , Bencilisoquinolinas/farmacología , Bencilisoquinolinas/uso terapéutico , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Janus Quinasa 2/efectos de los fármacos , Janus Quinasa 2/metabolismo , Pulmón/patología , Ratones Endogámicos BALB C , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico , Transducción de Señal , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
3.
J Complement Integr Med ; 20(2): 343-352, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-36935561

RESUMEN

OBJECTIVES: Obesity, diabetes mellitus, insulin resistance (IR), and hypertriglyceridemia are common features observed in non-alcoholic fatty liver diseases (NAFLD). There is a critical medical necessity to find novel therapeutics that can halt the development of NAFLD. METHODS: Bombax ceiba Linn. leaf extract was prepared and its phytochemical profile was determined. Standard and high carbohydrate high-fat diets (HCHF) were prepared. Rats were fed HCHF for 18 weeks to induce a non-alcoholic fatty liver (NAFL) model. Forty male rats were divided into control, B. ceiba Linn. leaf extract, NAFL, prophylactic, and treated groups. Serum fasting blood sugar (FBS), insulin, insulin resistance (HOMA-IR), cholesterol, high-density lipoprotein (HDL), triglycerides (TG), low density lipoprotein (LDL), alanine aminotransferase (ALT), aspartate aminotransferase (AST), intelectin-1 (ITLN1), p38 MAP kinase (MAPK), peroxisome proliferator-activated receptor alpha (PPAR-α), and interleukin-6 (IL-6) were evaluated. RESULTS: Data obtained showed that HCHF-induced NAFL resulting in a significant increase in FBS, serum insulin, HOMA-IR, cholesterol, LDL, TG, ALT, AST, and IL-6 and a significant decrease in serum levels of HDL, ITLN1, p38 MAP kinase, and PPAR-α compared to the control group. The analysis of B. ceiba Linn. leaf extract showed high content of phenol compounds which may cause a significant decrease in the levels of FBS, insulin, HOMA-IR values, lipid profile, and levels of IL-6 while a significant increase in serum levels of LDL, ITLN1, p38 MAP kinase, and PPAR-α compared to the NAFL group. CONCLUSIONS: B. ceiba Linn. Leaf extract is a highly protective and promising therapeutic agent against inflammation and oxidative stress in the NAFLD model induced by HCHF.


Asunto(s)
Bombax , Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Ratas , Masculino , Animales , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Hígado , Interleucina-6 , Triglicéridos , Insulina/uso terapéutico , Alanina Transaminasa , PPAR alfa/uso terapéutico , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Modelos Teóricos , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
4.
Int J Med Microbiol ; 313(2): 151576, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36812841

RESUMEN

INTRODUCTION: Pneumonia is an inflammation-related respiratory infection and chlorogenic acid (CGA) possesses a wide variety of bioactive properties, such as anti-inflammation and anti-bacteria. AIM: This study explored the anti-inflammatory mechanism of CGA in Klebsiella pneumoniae (Kp)-induced rats with severe pneumonia. METHODS: The pneumonia rat models were established by infection with Kp and treated with CGA. Survival rates, bacterial load, lung water content, and cell numbers in the bronchoalveolar lavage fluid were recorded, lung pathological changes were scored, and levels of inflammatory cytokines were determined by enzyme-linked immunosorbent assay. RLE6TN cells were infected with Kp and treated with CGA. The expression levels of microRNA (miR)-124-3p, p38, and mitogen-activated protein kinase (MAPK)-activated protein kinase 2 (MK2) in lung tissues and RLE6TN cells were quantified by real-time quantitative polymerase chain reaction or Western blotting. The binding of miR-124-3p to p38 was validated by the dual-luciferase and RNA pull-down assays. In vitro, the functional rescue experiments were performed using miR-124-3p inhibitor or p38 agonist. RESULTS: Kp-induced pneumonia rats presented high mortality, increased lung inflammatory infiltration and the release of inflammatory cytokines, and enhanced bacterial load, while CGA treatment improved rat survival rates and the above situations. CGA increased miR-124-3p expression, and miR-124-3p inhibited p38 expression and inactivated the p38MAPK pathway. Inhibition of miR-124-3p or activation of the p38MAPK pathway reversed the alleviative effect of CGA on pneumonia in vitro. CONCLUSION: CGA upregulated miR-124-3p expression and inactivated the p38MAPK pathway to downregulate inflammatory levels, facilitating the recovery of Kp-induced pneumonia rats.


Asunto(s)
MicroARNs , Neumonía , Ratas , Animales , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico , Klebsiella pneumoniae/genética , Ácido Clorogénico/farmacología , Ácido Clorogénico/uso terapéutico , Klebsiella/genética , Klebsiella/metabolismo , MicroARNs/genética , Neumonía/tratamiento farmacológico , Neumonía/microbiología , Citocinas/metabolismo , Antiinflamatorios/farmacología
5.
Planta Med ; 89(5): 484-492, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35789994

RESUMEN

To find inhibitors against skeletal muscle loss, we isolated a lignan compound ((-)-(2R,3R-1,4-O-diferuloylsecoisolarciresinol, DFS) from the stem of Alnus japonica. C2C12 myoblasts were treated with DFS during differentiation. To induce an in vitro atrophic condition, differentiated myotubes were treated with dexamethasone (a synthetic glucocorticoid). DFS (10 nM) increased expression levels of myogenic factors and the number of multi-nucleated myotubes expressing myosin heavy chain (MHC). The myogenic potential of DFS could be attributed to p38 MAPK activation. DFS also protected against dexamethasone-induced damage, showing increased expression of MHC and mammalian target of rapamycin (mTOR), a major anabolic factor. Under atrophic condition, the anti-myopathy effect of DFS was associated with inactivation of NF-κB signaling pathway and the subsequent suppression of muscle degradative E3 ligases and myostatin. DFS treatment also restored fast muscle fiber (type II a, II b, and II x), known to be susceptible to dexamethasone. These results indicate that DFS isolated from A. japonica can stimulate myogenesis via p38 MAPK activation and alleviate muscle atrophy by modulating the expression of genes associated with muscle protein anabolism/catabolism. Thus, we propose that DFS can be used as a pharmacological and nutraceutical agent for increasing muscle strength or protecting muscle loss.


Asunto(s)
Alnus , Lignanos , Alnus/metabolismo , Lignanos/farmacología , Músculo Esquelético/metabolismo , Atrofia Muscular/inducido químicamente , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/metabolismo , Fibras Musculares Esqueléticas , Dexametasona/efectos adversos , Desarrollo de Músculos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
6.
Basic Res Cardiol ; 117(1): 48, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36205817

RESUMEN

Although p38 MAP Kinase α (p38 MAPKα) is generally accepted to play a central role in the cardiac stress response, to date its function in maladaptive cardiac hypertrophy is still not unambiguously defined. To induce a pathological type of cardiac hypertrophy we infused angiotensin II (AngII) for 2 days via osmotic mini pumps in control and tamoxifen-inducible, cardiomyocyte (CM)-specific p38 MAPKα KO mice (iCMp38αKO) and assessed cardiac function by echocardiography, complemented by transcriptomic, histological, and immune cell analysis. AngII treatment after inactivation of p38 MAPKα in CM results in left ventricular (LV) dilatation within 48 h (EDV: BL: 83.8 ± 22.5 µl, 48 h AngII: 109.7 ± 14.6 µl) and an ectopic lipid deposition in cardiomyocytes, reflecting a metabolic dysfunction in pressure overload (PO). This was accompanied by a concerted downregulation of transcripts for oxidative phosphorylation, TCA cycle, and fatty acid metabolism. Cardiac inflammation involving neutrophils, macrophages, B- and T-cells was significantly enhanced. Inhibition of adipose tissue lipolysis by the small molecule inhibitor of adipocytetriglyceride lipase (ATGL) Atglistatin reduced cardiac lipid accumulation by 70% and neutrophil infiltration by 30% and went along with an improved cardiac function. Direct targeting of neutrophils by means of anti Ly6G-antibody administration in vivo led to a reduced LV dilation in iCMp38αKO mice and an improved systolic function (EF: 39.27 ± 14%). Thus, adipose tissue lipolysis and CM lipid accumulation augmented cardiac inflammation in iCMp38αKO mice. Neutrophils, in particular, triggered the rapid left ventricular dilatation. We provide the first evidence that p38 MAPKα acts as an essential switch in cardiac adaptation to PO by mitigating metabolic dysfunction and inflammation. Moreover, we identified a heart-adipose tissue-immune cell crosstalk, which might serve as new therapeutic target in cardiac pathologies.


Asunto(s)
Insuficiencia Cardíaca , Miocitos Cardíacos , Tejido Adiposo/metabolismo , Angiotensina II/metabolismo , Animales , Cardiomegalia/metabolismo , Ácidos Grasos/metabolismo , Inflamación/metabolismo , Lipasa/metabolismo , Lipasa/uso terapéutico , Lípidos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Neutrófilos/metabolismo , Tamoxifeno/metabolismo , Tamoxifeno/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
7.
Endocr Relat Cancer ; 29(12): 703-716, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36219868

RESUMEN

Prolactinomas have harmful effects on human health. Bromocriptine is the only commercially available drug in China, but about 25% of prolactinoma patients do not respond to it in clinic, its pathogenesis remains unknown. Thus, its pathogenesis needs to be determined to develop new therapeutic methods for prolactinomas. The expression of ERß, TLR4, and prolactin (PRL) in the pituitary gland of C57BL/6 mice and human prolactinoma specimen was examined by immunofluorescence or immunohistochemistry. The role of TLR4 in prolactinoma was determined using estradiol-induced models of C57BL/6 wild-type and TLR4-/- mice. MMQ cells were treated with estradiol, fulvestrant, and lipopolysaccharide (LPS) or transfected with TLR4 siRNA to study the expression of ERß, TLR4, and PRL in these cells. Furthermore, the interaction between ERß and TLR4 was investigated by immunoprecipitation analysis. The expression of PRL and TLR4 was co-located and increased in the pituitary gland of mice and human prolactinoma specimen compared to that in the control specimen. Meanwhile, TLR4 knockout or treatment with the TLR4 inhibitor TAK242 not only significantly inhibited tumor overgrowth but also decreased the expression of PRL in estradiol-treated mice through p38 MAPK pathway regulation. However, MMQ treated with estradiol and LPS enhanced PRL expression than treated with estradiol or LPS alone. Finally, ERß or TLR4 inhibition prevented the estradiol-induced PRL increase by regulating the TLR4/p38 MAPK pathway in vitro. Estradiol promoted prolactinoma development by activating the TLR4/p38 MAPK pathway through ERß, and TLR4 is a potential therapeutic target for prolactinoma treatment.


Asunto(s)
Neoplasias Hipofisarias , Prolactinoma , Animales , Humanos , Ratones , Estradiol/uso terapéutico , Receptor beta de Estrógeno , Estrógenos , Lipopolisacáridos , Ratones Endogámicos C57BL , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico , Neoplasias Hipofisarias/inducido químicamente , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/patología , Prolactina/metabolismo , Prolactinoma/inducido químicamente , Prolactinoma/tratamiento farmacológico , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/uso terapéutico
8.
Breast Cancer ; 29(6): 1076-1087, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35882754

RESUMEN

BACKGROUND: Sacituzumab govitecan is an antibody-drug conjugate that delivers SN-38, an active metabolite of irinotecan, to the target molecule, trophoblast cell-surface antigen 2 (Trop-2). It is a promising drug for triple-negative breast cancer and is anticipated to be effective for luminal breast cancer. The efficacy of the agent relies on the expression of Trop-2 rather than its intracellular function. However, conditions that alter the Trop-2 expression have not been well investigated. METHODS: We tested a range of clinically related treatments for their effect on Trop-2 expression in cultured breast cancer cell lines. RESULTS: The expression level of Trop-2 differed among cell lines, independent of their subtypes, and was highly variable on treatment with kinase inhibitors, tamoxifen, irradiation, and chemotherapeutic agents including irinotecan. While inhibitors of AKT, RSK, and p38 MAPK suppressed the Trop-2 expression, tamoxifen treatment significantly increased Trop-2 expression in luminal cancer cell lines. Notably, luminal cancer cells with acquired resistance to tamoxifen also exhibited higher levels of Trop-2. We identified transcription factor EB (TFEB) as a possible mechanism underlying tamoxifen-induced elevation of Trop-2 expression. Tamoxifen triggers dephosphorylation of TFEB, an active form of TFEB, and the effect of tamoxifen on Trop-2 was prevented by depletion of TFEB. A luciferase reporter assay showed that Trop-2 induction by TFEB was dependent on a tandem E-box motif within the Trop-2 promoter region. CONCLUSIONS: Overall, these results suggest that the effectiveness of sacituzumab govitecan could be altered by concomitant treatment and that tamoxifen could be a favorable agent for combined therapy.


Asunto(s)
Neoplasias de la Mama , Inmunoconjugados , Neoplasias de la Mama Triple Negativas , Femenino , Humanos , Antígenos de Neoplasias/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Camptotecina/farmacología , Inmunoconjugados/farmacología , Irinotecán/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico , Proteínas Proto-Oncogénicas c-akt , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Factores de Transcripción , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
9.
Contrast Media Mol Imaging ; 2022: 6551358, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35655729

RESUMEN

Nervous inflammation is an important component of the pathogenesis of neurodegenerative diseases including chronic diabetic neuropathic pain. In order to obtain a decrease in the progression of diabetic neuronal damage, it may be necessary to examine therapeutic options that involve antioxidants and anti-inflammatory agents. The aim of this study was to investigate the attenuation of inflammatory factors with endurance training in the spinal cord of rats with neuropathic pain. Thirty-two 8-week-old male Wistar rats (with a weight range of 204 ± 11.3 g) were randomly divided into 4 groups (n = 8), including (1) diabetic neuropathy (50 mg/kg streptozotocin intraperitoneal injection), (2) diabetic neuropathy training (30 minutes of endurance training at 15 meters per minute, 5 days a week for 6 weeks), (3) healthy training, and (4) healthy control. After confirmation of diabetic neuropathy by behavioral tests, training protocol and supplementation were performed. The NLRP3, P38 MAPK, TNF-α, and IL-1ß gene expressions were measured by a real-time technique in the spinal cord tissue. One-way analysis of variance and Tukey's post hoc test were used for statistical analysis. Endurance training reduced the sensitivity of the nervous system to thermal hyperalgesia and mechanical allodynia; also, compared to the diabetic neuropathy group, the gene expressions of NLTP3, P38 MAPK, TNF-α, and IL-1ß were significantly reduced by endurance training (P < 0.05). Endurance training modulates NLRP3, P38 MAPK, and TNF-α, IL-1ß gene expressions and improves the sensitivity of nociceptors to pain factors. Accordingly, it is recommended to use endurance training to reduce neuropathic pain for diabetics.


Asunto(s)
Diabetes Mellitus , Neuropatías Diabéticas , Entrenamiento Aeróbico , Neuralgia , Animales , Biomarcadores/metabolismo , Neuropatías Diabéticas/tratamiento farmacológico , Neuropatías Diabéticas/metabolismo , Neuropatías Diabéticas/patología , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Ratas , Ratas Wistar , Médula Espinal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
10.
Genes Genomics ; 44(7): 801-812, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35567715

RESUMEN

BACKGROUND: Tissue factor pathway inhibitor-1 (TFPI) is a serine protease inhibitor, which is responsible for inactivating TF-induced coagulation. Recently, increasing studies revealed that TFPI was lowly expressed in tumor cells and exhibited the antitumor activity. OBJECTIVE: The aim of this study was to explore the role and underlying molecular mechanisms of TFPI in breast cancer. METHODS: The expression and prognostic value of TFPI were analyzed using UALCAN and Kaplan-Meier plotter website. The expression level of TFPI in breast cancer tissues and cells was examined by immunohistochemistry (IHC) and western blot analysis, respectively. Cellular proliferation was evaluated by CCK-8 and colony formation assays. Cell migration and invasion were determined by transwell assay. The methylation level of TFPI promoter was determined by methylation-specific PCR. RESULTS: TFPI expression was significantly lower in breast cancer tissues and cells compared to normal breast tissues and normal breast cells. Patients with low TFPI levels showed worse overall survival (OS). Furthermore, overexpression of TFPI significantly inhibited the proliferation, migration and invasion of breast cancer cells. Conversely, knockdown of TFPI promoted the proliferation, migration and invasion of breast cancer cells. Mechanistically, TFPI inhibited the ERK/p38 MAPK signaling pathway in breast cancer. Moreover, DNA hypermethylation of TFPI promoter was responsible for the downregulation of TFPI in breast cancer cells. CONCLUSION: TFPI inhibited breast cancer cell proliferation, migration and invasion through inhibition of the ERK/p38 MAPK signaling pathway, suggesting that TFPI may serve as a novel prognostic biomarker and therapeutic target for breast cancer.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Lipoproteínas/genética , Lipoproteínas/metabolismo , Lipoproteínas/uso terapéutico , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
11.
Neurotherapeutics ; 19(5): 1634-1648, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35501470

RESUMEN

Despite the effort on developing new treatments, therapy for neuropathic pain is still a clinical challenge and combination therapy regimes of two or more drugs are often needed to improve efficacy. Accumulating evidence shows an altered expression and activity of histone acetylation enzymes in chronic pain conditions and restoration of these aberrant epigenetic modifications promotes pain-relieving activity. Recent studies showed a synergistic activity in neuropathic pain models by combination of histone deacetylases (HDACs) and bromodomain and extra-terminal domain (BET) inhibitors. On these premises, the present study investigated the pharmacological profile of new dual HDAC/BRD4 inhibitors, named SUM52 and SUM35, in the spared nerve injury (SNI) model in mice as innovative strategy to simultaneously inhibit HDACs and BETs. Intranasal administration of SUM52 and SUM35 attenuated thermal and mechanical hypersensitivity in the absence of locomotor side effects. Both dual inhibitors showed a preferential interaction with BRD4-BD2 domain, and SUM52 resulted the most active compound. SUM52 reduced microglia-mediated spinal neuroinflammation in spinal cord sections of SNI mice as showed by reduction of IBA1 immunostaining, inducible nitric oxide synthase (iNOS) expression, p65 nuclear factor-κB (NF-κB) and p38 MAPK over-phosphorylation. A robust decrease of the spinal proinflammatory cytokines content (IL-6, IL-1ß) was also observed after SUM52 treatment. Present results, showing the pain-relieving activity of HDAC/BRD4 dual inhibitors, indicate that the simultaneous modulation of BET and HDAC activity by a single molecule acting as multi-target agent might represent a promise for neuropathic pain relief.


Asunto(s)
Microglía , Neuralgia , Ratones , Animales , Microglía/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Nucleares/farmacología , Proteínas Nucleares/uso terapéutico , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Interleucina-6/metabolismo , Histonas/metabolismo , Factores de Transcripción , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Médula Espinal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico , Citocinas/metabolismo , Histona Desacetilasas/metabolismo , Histona Desacetilasas/farmacología , Histona Desacetilasas/uso terapéutico
12.
Am Heart J ; 243: 147-157, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34508693

RESUMEN

BACKGROUND: p38 mitogen activated kinase (MAPK) mediates the response to pro-inflammatory cytokines following myocardial infarction (MI) and is inhibited by losmapimod. METHODS: LATITUDE-TIMI 60 (ClinicalTrials.gov NCT02145468) randomized patients with MI to losmapimod or placebo for 12 weeks (24 weeks total follow-up). In this pre-specified analysis, we examined outcomes based on MI type [ST-segment elevation MI (STEMI) (865, 25%) and non-STEMI (2624, 75%)]. RESULTS: In patients with STEMI, inflammation, measured by hs-CRP, was significantly attenuated with losmapimod at 48 hours (P <0.001) and week 12 (P = 0.01). Losmapimod lowered NT-proBNP in patients with STEMI at 48 hours (P = 0.04) and week 12 (P = 0.02). The effects of losmapimod on CV death (CVD), MI, or severe recurrent ischemia requiring urgent coronary artery revascularization at 24 weeks [MACE] differed in patients with STEMI (7.0% vs 10.8%; HR 0.65, 95%CI 0.41 - 1.03; P= 0.06) and NSTEMI (11.4% vs 8.5%; HR 1.30, 95%CI 1.02 - 1.66; P = 0.04; p[int] = 0.009). CVD or HHF among patients with STEMI were 5.6% (losmapimod) and 8.3% (placebo) (HR 0.66; 95%CI 0.40 - 1.11; P = 0.12) and in NSTEMI were 4.8% (losmapimod) and 4.4% (placebo) (HR 1.09; 95%CI 0.76 - 1.56) in patients with NSTEMI. CONCLUSIONS: Patients with STEMI treated with losmapimod had an attenuated inflammatory response. Our collective findings raise the hypothesis that mitigating the inflammatory response may result in different outcomes in patients with STEMI and NSTEMI. While the difference in outcomes is exploratory, these findings do support separate examination of patients with STEMI and NSTEMI and increased emphasis on heart failure in future investigation of modulators of inflammation in MI.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Infarto del Miocardio sin Elevación del ST , Infarto del Miocardio con Elevación del ST , Humanos , Infarto del Miocardio/diagnóstico , Infarto del Miocardio con Elevación del ST/tratamiento farmacológico , Resultado del Tratamiento , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
13.
Adv Ther ; 39(1): 360-378, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34739698

RESUMEN

INTRODUCTION: Ovarian cancer has a high mortality rate due to difficulties in early detection and chemotherapy resistance. Human epididymal protein 4 (HE4) has been adopted as a novel serum biomarker for early ovarian cancer diagnosis, and the presence of Lewis y antigen modifications on HE4 in ovarian cancer cell lines has been detected in previous studies. The aim of this study was to analyze the expression of HE4 and Lewis y antigen in human ovarian cancer in order to find a correlation between them, as well as with the clinical pathological parameters of patients with ovarian cancer. METHODS: Immunohistochemistry was used to detect the respective expression of these compounds in two patient groups (chemotherapy-resistant and chemotherapy-sensitive) containing a total of 95 patients. Then, a bioinformatic approach was adopted and online large sample databases (TCGA, CCLE, and GTEx; Metascape, Cytoscape) were used to explore the potential mechanisms of action of these compounds. RESULTS: The results of this study demonstrate that high HE4 and Lewis y expression could be used as markers for chemotherapy resistance and poor prognosis in patients with ovarian cancer. These two expression events were widely correlated in various cancer tissues and are thought to act by activating the p38 mitogen-activated protein kinases (MAPK) pathway and inducing Vascular Endothelial Growth Factor A (VEGFA), Prostaglandin-Endoperoxide Synthase 2 (PTGS2), Early Growth Response 1 (EGR1), and Hypoxia-Inducible Factor 1-Alpha (HIFI1A), thereby promoting malignant biological behavior and resistance in ovarian cancer. CONCLUSIONS: These findings not only reveal the possible mechanism by which HE4 and Lewis y antigen affect ovarian cancer but also identify a four-gene signature that could be very useful in ovarian cancer detection and/or the development of new targeted therapies.


Asunto(s)
Neoplasias Glandulares y Epiteliales , Neoplasias Ováricas , Biomarcadores de Tumor/metabolismo , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Humanos , Inmunohistoquímica , Neoplasias Ováricas/patología , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
14.
J Recept Signal Transduct Res ; 41(2): 153-158, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32752909

RESUMEN

MAP kinase is one of the important targets in the treatment of osteoarthritis, inflammation and cancer. Many p38 inhibitors with diverse chemical structures and modes of protein interaction have been designed on the basis of their ability to compete with ATP site or allosteric site for binding to MAP Kinase. This study involves the molecular docking of benzimidazoles containing 4H-chrome-4-one derivatives as potent inhibitors of the MAP kinase enzyme. The compounds were computationally designed and optimized with the molecular docking to investigate the interactions between the target compounds and the amino acid residues of the MAP Kinase. The inhibitory activities against human MAP kinase enzyme were investigated by molecular docking using the Autodock and discovery studio software. All the designed compounds were shown good binding energy when compared with the binging energies of standard drug Imatinib (anti-cancer). Among all the designed compounds, compound D1 and D6 have higher binding energy values when compared to standard drug. Here we also studied the molecular properties of designed compound using Molinspiration software. Further, we planned to synthesis these benzimidazole derivatives and screen for in-vitro and in-vivo of anti-cancer activity.


Asunto(s)
Bencimidazoles/química , Diseño de Fármacos , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas p38 Activadas por Mitógenos/química , Adenosina Trifosfato/química , Sitio Alostérico/efectos de los fármacos , Bencimidazoles/uso terapéutico , Simulación por Computador , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Mesilato de Imatinib , Simulación del Acoplamiento Molecular , Estructura Molecular , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Relación Estructura-Actividad , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/ultraestructura
15.
Clinics (Sao Paulo) ; 74: e509, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31411275

RESUMEN

Acute respiratory distress syndrome (ARDS) is a life-threatening illness characterized by a complex pathophysiology, involving not only the respiratory system but also nonpulmonary distal organs. Although advances in the management of ARDS have led to a distinct improvement in ARDS-related mortality, ARDS is still a life-threatening respiratory condition with long-term consequences. A better understanding of the pathophysiology of this condition will allow us to create a personalized treatment strategy for improving clinical outcomes. In this article, we present a general overview p38 mitogen-activated protein kinase (p38MAPK) and recent advances in understanding its functions. We consider the potential of the pharmacological targeting of p38MAPK pathways to treat ARDS.


Asunto(s)
Mediadores de Inflamación/metabolismo , Síndrome de Dificultad Respiratoria/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
16.
Clinics ; 74: e509, 2019.
Artículo en Inglés | LILACS | ID: biblio-1011922

RESUMEN

Acute respiratory distress syndrome (ARDS) is a life-threatening illness characterized by a complex pathophysiology, involving not only the respiratory system but also nonpulmonary distal organs. Although advances in the management of ARDS have led to a distinct improvement in ARDS-related mortality, ARDS is still a life-threatening respiratory condition with long-term consequences. A better understanding of the pathophysiology of this condition will allow us to create a personalized treatment strategy for improving clinical outcomes. In this article, we present a general overview p38 mitogen-activated protein kinase (p38MAPK) and recent advances in understanding its functions. We consider the potential of the pharmacological targeting of p38MAPK pathways to treat ARDS.


Asunto(s)
Humanos , Síndrome de Dificultad Respiratoria del Recién Nacido/fisiopatología , Mediadores de Inflamación/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/inmunología , Síndrome de Dificultad Respiratoria del Recién Nacido/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico , Inflamación/inmunología , Inflamación/metabolismo
17.
Neurosci Lett ; 581: 89-93, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25172145

RESUMEN

The dedicator of cytokinesis 3 (Dock3) is an atypical guanine nucleotide exchange factor that is predominantly expressed in the CNS. Dock3 exerts neuroprotective effects and stimulates optic nerve regeneration. The p38 mitogen-activated protein kinase acts downstream of apoptosis signal-regulating kinase 1 (ASK1) signaling and plays an important role in neural cell death. We assessed a therapeutic efficacy of Dock3 stimulation and p38 inhibition in retinal degeneration induced by optic nerve injury (ONI). In vivo retinal imaging using optical coherence tomography revealed that ONI-induced retinal degeneration was ameliorated in SB203580 (a p38 inhibitor)-treated WT mice and PBS-treated Dock3 overexpressing (Dock3 Tg) mice, and SB203580 further stimulated retinal protection in Dock3 Tg mice. In addition, SB203580 increased the number of regenerating axons after ONI in both WT and Dock3 Tg mice. ONI-induced phosphorylation of ASK1, p38 and the N-methyl-d-aspartate receptor 2B subunit were suppressed in the retina of Dock3 Tg mice. Inhibition of the ASK1 pathway in Dock3 Tg mice suggests that Dock3 may have an antioxidant-like property. These results indicate that overexpression of Dock3 and pharmacological interruption of p38 have synergistic effects for both neuroprotection and axon regeneration, thus combined application may be beneficial for the treatment of ONI.


Asunto(s)
Axones/fisiología , Proteínas Portadoras/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Traumatismos del Nervio Óptico/metabolismo , Regeneración/fisiología , Células Ganglionares de la Retina/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Proteínas Portadoras/uso terapéutico , Recuento de Células , Inhibidores Enzimáticos/farmacología , Factores de Intercambio de Guanina Nucleótido , Imidazoles/farmacología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/uso terapéutico , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Traumatismos del Nervio Óptico/enzimología , Traumatismos del Nervio Óptico/terapia , Fosforilación , Piridinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
18.
Expert Opin Investig Drugs ; 18(12): 1893-905, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19852565

RESUMEN

BACKGROUND: The p38 mitogen-activated protein kinase (MAPK) is activated by various pro-inflammatory and stressful stimuli. Mounting evidence suggests that the p38 MAPK signaling cascade is involved in various biological responses other than inflammation such as cell proliferation, differentiation, apoptosis and invasion, suggesting that the p38 MAPK can serve as a potential therapeutic target for the treatment of not only inflammatory diseases but also cancer. METHODS: The unique characteristics of p38 MAPK are summarized with regard to activation and function of p38 MAPK signaling cascades. We then discuss the involvement of p38 MAPK in diseases and the implications of the possible therapeutic use of p38 MAPK inhibitors. The p38 MAPK inhibitors that have been used in the in vitro/in vivo systems as well as in the clinical trials are summarized. RESULTS/CONCLUSION: The p38 MAPK plays an important role in key cellular processes related to inflammation and cancer. Understanding the signal transduction mechanisms and gene regulation by p38 MAPK provides useful information in the development of p38 MAPK inhibitors with therapeutic benefits with reduced side effects. In this review, we summarize and present the list of p38 MAPK inhibitors in in vitro/in vivo studies as well as in clinical trials.


Asunto(s)
Drogas en Investigación/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Inflamación/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Drogas en Investigación/farmacología , Humanos , Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Drug News Perspect ; 17(9): 607-14, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15645018

RESUMEN

The 228th American Chemical Society (ACS) National Meeting was held August 22-26, 2004, in Philadelphia, Pennsylvania. More than 400 medicinal chemistry papers were presented, most in symposia, general sessions and poster sessions sponsored by the ACS Division of Medicinal Chemistry. One major focus of the meeting was the development of novel drugs for the treatment of inflammatory and autoimmune diseases such as rheumatoid arthritis, asthma, inflammatory bowel disease (Crohn's disease), psoriasis, reperfusion injury and endotoxic shock. This report will center on said areas of research.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Química Farmacéutica/tendencias , Animales , Enfermedades Autoinmunes/inmunología , Química Farmacéutica/métodos , Química Farmacéutica/organización & administración , Quimiocinas/antagonistas & inhibidores , Quimiocinas/inmunología , Quimiocinas/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Humanos , Hidrazonas , Interleucinas/antagonistas & inhibidores , Interleucinas/inmunología , Interleucinas/uso terapéutico , Morfolinas/antagonistas & inhibidores , Morfolinas/inmunología , Morfolinas/uso terapéutico , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/uso terapéutico , Philadelphia , Pirimidinas , Receptores Tipo II del Factor de Necrosis Tumoral , Triazinas/antagonistas & inhibidores , Triazinas/inmunología , Triazinas/uso terapéutico , Receptores Señuelo del Factor de Necrosis Tumoral , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...