Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.428
Filtrar
1.
Anal Chem ; 96(26): 10724-10731, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38952276

RESUMEN

Carboxylesterase (CE), an enzyme widely present in organisms, is involved in various physiological and pathological processes. Changes in the levels of CEs in the liver may predict the presence of type 2 diabetes mellitus (T2DM). Here, a novel dicyanoisophorone (DCI)-based proximity-labeled far-red fluorescent probe DCI2F-Ac with endoplasmic reticulum targeting was proposed for real-time monitoring and imaging of the CEs activity. DCI2F-Ac featured very low cytotoxicity and biotoxicity and was highly selective and sensitive for CEs. Compared with traditional CEs probes, DCI2F-Ac was covalently anchored directly to CEs, thus effectively reducing the loss of in situ fluorescent signals due to diffusion. Through the "on-off" fluorescence signal readout, DCI2F-Ac was able to distinguish cell lines and screen for CEs inhibitors. In terms of endoplasmic reticulum (ER) stress, it was found that thapsigargin (Tg) induced upregulation of CEs levels but not tunicamycin (Tm), which was related to the calcium homeostasis of the ER. DCI2F-Ac could efficiently detect downregulated CEs in the livers of T2DM, and the therapeutic efficacy of metformin, acarbose, and a combination of these two drugs was assessed by tracking the fluctuation of CEs levels. The results showed that combining metformin and acarbose could restore CEs levels to near-normal levels with the best antidiabetic effect. Thus, the DCI2F-Ac probe provides a great opportunity to explore the untapped potential of CEs in liver metabolic disorders and drug efficacy assessment.


Asunto(s)
Carboxilesterasa , Diabetes Mellitus Tipo 2 , Retículo Endoplásmico , Colorantes Fluorescentes , Colorantes Fluorescentes/química , Colorantes Fluorescentes/síntesis química , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Carboxilesterasa/metabolismo , Carboxilesterasa/antagonistas & inhibidores , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Animales , Ratones , Imagen Óptica , Células Hep G2 , Estrés del Retículo Endoplásmico/efectos de los fármacos
2.
Mol Biol (Mosk) ; 58(1): 141-153, 2024.
Artículo en Ruso | MEDLINE | ID: mdl-38943585

RESUMEN

Melatonin (N-acetyl-5-methoxytryptamine, MEL) is a hormone synthesized by the pineal gland. Due to its oncostatic effect, it can be considered as an antitumor agent and used for combination therapy. ABT-737, a Bcl-2 inhibitor, promotes cell death after treatment with agents that induce pro-apoptotic signals. In the present study, the combined effect of MEL and ABT-737 on changes in proliferative and mitotic activity, mitochondrial membrane potential, intracellular production of reactive oxygen species (ROS), and cytosolic Ca^(2+) was studied. Moreover, changes in the expression of anti- and pro-apoptotic proteins (Bcl-2 and Bax), autophagy markers (LC3A/B (I, II)), endoplasmic reticulum stress markers (chaperones BIP and PDI, CHOP) were studied under these conditions. The effect of MEL together with ABT-737 led to an increase in the level of cytosolic Ca^(2+), intracellular production of ROS and a decrease in the membrane potential of mitochondria. The content of Bcl-2 increased, while the level of Bax decreased. Activation of CHOP stimulated autophagy and led to a decrease in the synthesis of chaperones BIP and PDI. It is assumed that melatonin can enhance the effect of other chemotherapeutic agents and can be used in the treatment of tumors.


Asunto(s)
Apoptosis , Compuestos de Bifenilo , Melatonina , Potencial de la Membrana Mitocondrial , Nitrofenoles , Piperazinas , Proteínas Proto-Oncogénicas c-bcl-2 , Especies Reactivas de Oxígeno , Sulfonamidas , Humanos , Sulfonamidas/farmacología , Melatonina/farmacología , Nitrofenoles/farmacología , Piperazinas/farmacología , Compuestos de Bifenilo/farmacología , Especies Reactivas de Oxígeno/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Células THP-1 , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Sinergismo Farmacológico , Autofagia/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Proliferación Celular/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Calcio/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/biosíntesis , Factor de Transcripción CHOP
3.
J Toxicol Sci ; 49(7): 313-319, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38945842

RESUMEN

Dihydropyrazines (DHPs) are formed by non-enzymatic glycation reactions in vivo and in food. We recently reported that 3-hydro-2,2,5,6-tetramethylpyrazine (DHP-3), which is a methyl-substituted DHP, caused severe oxidative stress and cytotoxicity. However, the molecular mechanisms underlying the cytotoxic pathways of the DHP response remain elusive. Because oxidative stress induces endoplasmic reticulum (ER) stress and autophagy, we investigated the ability of DHP-3 to modulate the ER stress and autophagy pathways. DHP-3 activated the ER stress pathway by increasing inositol-requiring enzyme 1 (IRE1) and PKR-like ER kinase (PERK) phosphorylation and transcription factor 6 (ATF6) expression. Moreover, DHP-3 increased the expression of activating transcription factor 4 (ATF4) and C/EBP homologous protein (CHOP), which are downstream targets of PERK. In addition, DHP-3 inhibited the autophagy pathway by increasing the accumulation of microtubule-associated protein 1 light chain 3 alpha-phosphatidylethanolamine conjugate (LC3-II) and p62/sequestosome 1 (p62), while decreasing autophagic flux. Taken together, these results indicate that DHP-3 activates the ER stress pathway and inhibits the autophagy pathway, suggesting that the resulting removal of damaged organelles is inadequate.


Asunto(s)
Factor de Transcripción Activador 4 , Factor de Transcripción Activador 6 , Autofagia , Estrés del Retículo Endoplásmico , Proteínas Serina-Treonina Quinasas , Pirazinas , eIF-2 Quinasa , Humanos , Autofagia/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Pirazinas/farmacología , Células Hep G2 , Factor de Transcripción Activador 4/metabolismo , Factor de Transcripción Activador 4/genética , eIF-2 Quinasa/metabolismo , Factor de Transcripción Activador 6/metabolismo , Factor de Transcripción Activador 6/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Fosforilación , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Estrés Oxidativo/efectos de los fármacos , Proteína Sequestosoma-1/metabolismo , Proteína Sequestosoma-1/genética , Transducción de Señal/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo
4.
Vet Res ; 55(1): 78, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877574

RESUMEN

Endoplasmic reticulum stress (ERS) and oxidative stress (OS) are adaptive responses of the body to stressor stimulation. Although it has been verified that Trichinella spiralis (T. spiralis) can induce ERS and OS in the host, their association is still unclear. Therefore, this study explored whether T. spiralis-secreted serpin-type serine protease inhibitor (TsAdSPI) is involved in regulating the relationship between ERS and OS in the host intestine. In this study, mice jejunum and porcine small intestinal epithelial cells (IECs) were detected using qPCR, western blotting, immunohistochemistry (IHC), immunofluorescence (IF), and detection kits. The results showed that ERS- and OS-related indexes changed significantly after TsAdSPI stimulation, and Bip was located in IECs, indicating that TsAdSPI could induce ERS and OS in IECs. After the use of an ERS inhibitor, OS-related indexes were inhibited, suggesting that TsAdSPI-induced OS depends on ERS. When the three ERS signalling pathways, ATF6, IRE1, and PERK, were sequentially suppressed, OS was only regulated by the PERK pathway, and the PERK-eif2α-CHOP-ERO1α axis played a key role. Similarly, the expression of ERS-related indexes and the level of intracellular Ca2+ were inhibited after adding the OS inhibitor, and the expression of ERS-related indexes decreased significantly after inhibiting calcium transfer. This finding indicated that TsAdSPI-induced OS could affect ERS by promoting Ca2+ efflux from the endoplasmic reticulum. The detection of the ERS and OS sequences revealed that OS occurred before ERS. Finally, changes in apoptosis-related indexes were detected, and the results indicated that TsAdSPI-induced ERS and OS could regulate IEC apoptosis. In conclusion, TsAdSPI induced OS after entering IECs, OS promoted ERS by enhancing Ca2+ efflux, and ERS subsequently strengthened OS by activating the PERK-eif2α-CHOP-ERO1α axis. ERS and OS induced by TsAdSPI synergistically promoted IEC apoptosis. This study provides a foundation for exploring the invasion mechanism of T. spiralis and the pathogenesis of host intestinal dysfunction after invasion.


Asunto(s)
Estrés del Retículo Endoplásmico , Células Epiteliales , Estrés Oxidativo , Serpinas , Trichinella spiralis , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Trichinella spiralis/fisiología , Ratones , Estrés Oxidativo/efectos de los fármacos , Porcinos , Serpinas/metabolismo , Serpinas/genética , Inhibidores de Serina Proteinasa/farmacología , Proteínas del Helminto/metabolismo , Proteínas del Helminto/genética , Yeyuno/efectos de los fármacos
5.
ACS Appl Mater Interfaces ; 16(26): 33169-33181, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38915234

RESUMEN

Inducing immunogenic cell death (ICD) is a promising strategy for cancer immunotherapy. Shikonin (SHK), a naphthoquinone compound from Lithospermum erythrorhizon, can stimulate antitumor immunity by inducing ICD. Nevertheless, the immunogenicity of tumor cells killed by SHK is weak. Endoplasmic reticulum (ER) stress is an important intracellular pathway of the ICD effect. Curcumin (CUR) can directly induce ER stress by disrupting Ca2+ homeostasis, which might enhance SHK-induced ICD effect. A self-delivery ICD effect nanobooster (CS-PEG NPs) was developed by the self-assembly of SHK (ICD inducer) and CUR (ICD enhancer) with the assistance of DSPE-PEG2K for cancer chemoimmunotherapy. CS-PEG NPs possessed effective CT26 tumor cell cellular uptake and tumor accumulation ability. Moreover, enhanced cytotoxicity against tumor cells and apoptosis promotion were achieved due to the synergistic effect of CUR and SHK. Notably, CS-PEG NPs induced obvious Ca2+ homeostasis disruption, ER stress, and ICD effect. Subsequently, the neoantigens produced by the robust ICD effect in vivo promoted dendritic cell maturation, which further recruited and activated cytotoxic T lymphocytes. Superior antitumor efficacy and systemic antitumor immunity were observed in the CT26-bearing BALB/c mouse model without side effects in major organs. This study offers a promising self-delivery nanobooster to induce strong ICD effect and antitumor immunity for cancer chemoimmunotherapy.


Asunto(s)
Curcumina , Estrés del Retículo Endoplásmico , Muerte Celular Inmunogénica , Inmunoterapia , Ratones Endogámicos BALB C , Naftoquinonas , Animales , Naftoquinonas/química , Naftoquinonas/farmacología , Muerte Celular Inmunogénica/efectos de los fármacos , Ratones , Curcumina/química , Curcumina/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Línea Celular Tumoral , Nanopartículas/química , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/patología , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Femenino
6.
Nat Aging ; 4(6): 839-853, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38858606

RESUMEN

Thermogenic beige adipocytes are recognized as potential therapeutic targets for combating metabolic diseases. However, the metabolic advantages that they offer are compromised with aging. Here we show that treating mice with estrogen (E2), a hormone that decreases with age, can counteract the age-related decline in beige adipogenesis when exposed to cold temperature while concurrently enhancing energy expenditure and improving glucose tolerance in mice. Mechanistically, we found that nicotinamide phosphoribosyl transferase (NAMPT) plays a pivotal role in facilitating the formation of E2-induced beige adipocytes, which subsequently suppresses the onset of age-related endoplasmic reticulum (ER) stress. Furthermore, we found that targeting NAMPT signaling, either genetically or pharmacologically, can restore the formation of beige adipocytes by increasing the number of perivascular adipocyte progenitor cells. Conversely, the absence of NAMPT signaling prevents this process. Together, our findings shed light on the mechanisms regulating the age-dependent impairment of beige adipocyte formation and underscore the E2-NAMPT-controlled ER stress pathway as a key regulator of this process.


Asunto(s)
Adipocitos Beige , Adipogénesis , Envejecimiento , Estrés del Retículo Endoplásmico , Estrógenos , Nicotinamida Fosforribosiltransferasa , Nicotinamida Fosforribosiltransferasa/metabolismo , Animales , Adipogénesis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ratones , Envejecimiento/efectos de los fármacos , Envejecimiento/fisiología , Estrógenos/metabolismo , Estrógenos/farmacología , Adipocitos Beige/efectos de los fármacos , Adipocitos Beige/metabolismo , Citocinas/metabolismo , Transducción de Señal/efectos de los fármacos , Femenino , Ratones Endogámicos C57BL , Metabolismo Energético/efectos de los fármacos
7.
Food Res Int ; 188: 114508, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38823847

RESUMEN

Procyanidins, which are oligomerized flavan-3-ols with a polyphenolic structure, are bioactive substances that exhibit various biological effects. However, the relationship between the degree of polymerization (DP) of procyanidins and their bioactivities remains largely unknown. In this study, the preventive effects of procyanidins with different DP (EC, PB2 and PC1) on glucose improvement and liver lipid deposition were investigated using a high-fat diet/streptozotocin-induced diabetes mouse model. The results demonstrated that all the procyanidins with different DP effectively reduced fasting blood glucose and glucose/insulin tolerance, decreased the lipid profile (total cholesterol, triglyceride, and low-density lipoprotein cholesterol content) in serum and liver tissue as well as the liver oil red staining, indicating the improvement of glucose metabolism, insulin sensitivity and hepatic lipid deposition in diabetic mice. Furthermore, the procyanidins down-regulated expression of glucose regulated 78-kDa protein (GRP78) and C/EBP homologous protein (CHOP), indicating a regulation role of endoplasmic reticulum (ER) stress. The inhibition of ER stress by tauroursodeoxycholic acid (TUDCA) treatment abolished the effects of procyanidins with different DP in PA-induced HepG2 cells, confirming that procyanidins alleviate liver hyperlipidemia through the modulation of ER stress. Molecular docking results showed that EC and PB2 could better bind GRP78 and CHOP. Collectively, our study reveals that the structure of procyanidins, particularly DP, is not directly correlated with the improvement of blood glucose and lipid deposition, while highlighting the important role of ER stress in the bioactivities of procyanidins.


Asunto(s)
Glucemia , Diabetes Mellitus Experimental , Dieta Alta en Grasa , Chaperón BiP del Retículo Endoplásmico , Metabolismo de los Lípidos , Hígado , Proantocianidinas , Animales , Proantocianidinas/farmacología , Dieta Alta en Grasa/efectos adversos , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Masculino , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Glucemia/metabolismo , Glucemia/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Células Hep G2 , Humanos , Polimerizacion , Estrés del Retículo Endoplásmico/efectos de los fármacos , Simulación del Acoplamiento Molecular , Biflavonoides/farmacología , Ratones Endogámicos C57BL , Estreptozocina , Resistencia a la Insulina , Catequina/farmacología
8.
J Transl Med ; 22(1): 538, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844946

RESUMEN

Apalutamide, a novel endocrine therapy agent, has been shown to significantly improve the prognosis of patients with metastatic hormone-sensitive prostate cancer (mHSPC). However, resistance to apalutamide has also been reported, and the underlying mechanism for this response has yet to be clearly elucidated. First, this study established apalutamide-resistant prostate cancer (PCa) cells, and confirmed that apalutamide activated the release of calcium ions (Ca2+) and endoplasmic reticulum stress (ERS) to enhance autophagy. Second, RNA sequencing, western blotting, and immunohistochemistry revealed significantly decreased Calpain 2 (CAPN2) expression in the apalutamide-resistant PCa cells and tissues. Furthermore, immunofluorescence and transmission electron microscopy (TEM) showed that CAPN2 promoted apalutamide resistance by activating protective autophagy. CAPN2 promoted autophagy by reducing Forkhead Box O1 (FOXO1) degradation while increasing nuclear translocation via nucleoplasmic protein isolation and immunofluorescence. In addition, FOXO1 promoted protective autophagy through the transcriptional regulation of autophagy-related gene 5 (ATG5). Furthermore, a dual-fluorescence assay confirmed that transcription factor 3 (ATF3) stimulation promoted CAPN2-mediated autophagy activation via transcriptional regulation. In summary, CAPN2 activated protective autophagy by inhibiting FOXO1 degradation and promoting its nuclear translocation via transcriptional ATG5 regulation. ATF3 activation and transcriptional CAPN2 regulation jointly promoted this bioeffect. Thus, our findings have not only revealed the mechanism underlying apalutamide resistance, but also provided a promising new target for the treatment of metastatic PCa.


Asunto(s)
Autofagia , Calpaína , Resistencia a Antineoplásicos , Metástasis de la Neoplasia , Neoplasias de la Próstata , Tiohidantoínas , Humanos , Masculino , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Calpaína/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Tiohidantoínas/farmacología , Tiohidantoínas/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Calcio/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Animales
9.
Sci Rep ; 14(1): 13116, 2024 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849435

RESUMEN

Stearoyl-CoA desaturase 1 (SCD1) is an attractive target for cancer therapy. However, the clinical efficacy of SCD1 inhibitor monotherapy is limited. There is thus a need to elucidate the mechanisms of resistance to SCD1 inhibition and develop new therapeutic strategies for combination therapy. In this study, we investigated the molecular mechanisms by which cancer cells acquire resistance to endoplasmic reticulum (ER) stress-dependent cancer cell death induced by SCD1 inhibition. SCD1 inhibitor-sensitive and -resistant cancer cells were treated with SCD1 inhibitors in vitro, and SCD1 inhibitor-sensitive cancer cells accumulated palmitic acid and underwent ER stress response-induced cell death. Conversely, SCD1-resistant cancer cells did not undergo ER stress response-induced cell death because fatty acid desaturase 2 (FADS2) eliminated the accumulation of palmitic acid. Furthermore, genetic depletion using siRNA showed that FADS2 is a key determinant of sensitivity/resistance of cancer cells to SCD1 inhibitor. A549 cells, an SCD1 inhibitor-resistant cancer cell line, underwent ER stress-dependent cancer cell death upon dual inhibition of SCD1 and FADS2. Thus, combination therapy with SCD1 inhibition and FADS2 inhibition is potentially a new cancer therapeutic strategy targeting fatty acid metabolism.


Asunto(s)
Resistencia a Antineoplásicos , Estrés del Retículo Endoplásmico , Ácido Graso Desaturasas , Estearoil-CoA Desaturasa , Estearoil-CoA Desaturasa/metabolismo , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Humanos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Ácido Graso Desaturasas/genética , Ácido Graso Desaturasas/metabolismo , Línea Celular Tumoral , Células A549 , Ácido Palmítico/farmacología , Muerte Celular/efectos de los fármacos , Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología , Neoplasias/tratamiento farmacológico
10.
Vet Med Sci ; 10(4): e1504, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38879885

RESUMEN

BACKGROUND: The metabolic impacts of including soya meal, wheat gluten and corn gluten in the diet of male lambs could influence their reproductive performance. OBJECTIVES: An experiment was carried out to assess the effects of corn gluten, wheat gluten and soya meal on the reproductive system of male lambs. METHODS: Twenty-four male Morkaraman lambs, aged 9 months, were utilized in this study and were fed experimental diets for 56 days. The lambs were divided into a control group (soybean meal + safflower meal), a corn group (corn gluten) and a wheat group (wheat gluten). RESULTS: The serum follicle-stimulating hormone level of the control group was significantly higher and tumour necrosis factor-alpha (TNF-α) level was lower than the wheat and corn gluten groups (p < 0.05). The lowest malondialdehyde level in testicular tissue was observed in the control group, whereas the highest was in the wheat gluten group (p < 0.05). The glutathione level in the control group was significantly higher than in the other groups (p < 0.05). The corn gluten group showed the highest CHOP and IRE1 levels; the lowest Bcl-2 levels and the highest IL-1B and P2 × 7R levels were found in the wheat group; and the lowest TNF-α levels were in the control group (p < 0.05). Additionally, the study revealed that diet had a significant impact on spermatological parameters of the testis such as diameter, volume and weight (p < 0.05). CONCLUSIONS: These results concluded that the inclusion of different protein sources in the diet of reproductive male lambs affects the metabolism of testicular tissue.


Asunto(s)
Alimentación Animal , Dieta , Estrés del Retículo Endoplásmico , Espermatozoides , Testículo , Animales , Masculino , Dieta/veterinaria , Alimentación Animal/análisis , Estrés del Retículo Endoplásmico/efectos de los fármacos , Espermatozoides/fisiología , Espermatozoides/efectos de los fármacos , Análisis de Semen/veterinaria , Oveja Doméstica/fisiología , Ovinos/fisiología , Triticum/química , Fenómenos Fisiológicos Nutricionales de los Animales , Zea mays/química , Glycine max/química
11.
Int J Mol Sci ; 25(11)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38891776

RESUMEN

Neural tube defects (NTDs), which are caused by impaired embryonic neural tube closure, are one of the most serious and common birth defects. Peptidyl-prolyl cis/trans isomerase 1 (Pin1) is a prolyl isomerase that uniquely regulates cell signaling by manipulating protein conformation following phosphorylation, although its involvement in neuronal development remains unknown. In this study, we explored the involvement of Pin1 in NTDs and its potential mechanisms both in vitro and in vivo. The levels of Pin1 expression were reduced in NTD models induced by all-trans retinoic acid (Atra). Pin1 plays a significant role in regulating the apoptosis, proliferation, differentiation, and migration of neurons. Moreover, Pin1 knockdown significantly was found to exacerbate oxidative stress (OS) and endoplasmic reticulum stress (ERs) in neuronal cells. Further studies showed that the Notch1-Nrf2 signaling pathway may participate in Pin1 regulation of NTDs, as evidenced by the inhibition and overexpression of the Notch1-Nrf2 pathway. In addition, immunofluorescence (IF), co-immunoprecipitation (Co-IP), and GST pull-down experiments also showed that Pin1 interacts directly with Notch1 and Nrf2. Thus, our study suggested that the knocking down of Pin1 promotes NTD progression by inhibiting the activation of the Notch1-Nrf2 signaling pathway, and it is possible that this effect is achieved by disrupting the interaction of Pin1 with Notch1 and Nrf2, affecting their proteostasis. Our research identified that the regulation of Pin1 by retinoic acid (RA) and its involvement in the development of NTDs through the Notch1-Nrf2 axis could enhance our comprehension of the mechanism behind RA-induced brain abnormalities.


Asunto(s)
Peptidilprolil Isomerasa de Interacción con NIMA , Defectos del Tubo Neural , Receptor Notch1 , Tretinoina , Tretinoina/metabolismo , Tretinoina/farmacología , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Animales , Ratones , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/inducido químicamente , Receptor Notch1/metabolismo , Receptor Notch1/genética , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Transducción de Señal/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Femenino , Tubo Neural/metabolismo , Tubo Neural/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Humanos
12.
Int J Mol Sci ; 25(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892135

RESUMEN

Podophyllotoxin (PPT) is an active pharmaceutical ingredient (API) with established antitumor potential. However, due to its systemic toxicity, its use is restricted to topical treatment of anogenital warts. Less toxic PPT derivatives (e.g., etoposide and teniposide) are used intravenously as anticancer agents. PPT has been exploited as a scaffold of new potential therapeutic agents; however, fewer studies have been conducted on the parent molecule than on its derivatives. We have undertaken a study of ultrastructural changes induced by PPT on HaCaT keratinocytes. We have also tracked the intracellular localization of PPT using its fluorescent derivative (PPT-FL). Moreover, we performed molecular docking of both PPT and PPT-FL to compare their affinity to various binding sites of tubulin. Using the Presto blue viability assay, we established working concentrations of PPT in HaCaT cells. Subsequently, we have used selected concentrations to determine PPT effects at the ultrastructural level. Dynamics of PPT distribution by confocal microscopy was performed using PPT-FL. Molecular docking calculations were conducted using Glide. PPT induces a time-dependent cytotoxic effect on HaCaT cells. Within 24 h, we observed the elongation of cytoplasmic processes, formation of cytoplasmic vacuoles, progressive ER stress, and shortening of the mitochondrial long axis. After 48 h, we noticed disintegration of the cell membrane, progressive vacuolization, apoptotic/necrotic vesicles, and a change in the cell nucleus's appearance. PPT-FL was detected within HaCaT cells after ~10 min of incubation and remained within cells in the following measurements. Molecular docking confirmed the formation of a stable complex between tubulin and both PPT and PPT-FL. However, it was formed at different binding sites. PPT is highly toxic to normal human keratinocytes, even at low concentrations. It promptly enters the cells, probably via endocytosis. At lower concentrations, PPT causes disruptions in both ER and mitochondria, while at higher concentrations, it leads to massive vacuolization with subsequent cell death. The novel derivative of PPT, PPT-FL, forms a stable complex with tubulin, and therefore, it is a useful tracker of intracellular PPT binding and trafficking.


Asunto(s)
Células HaCaT , Queratinocitos , Simulación del Acoplamiento Molecular , Podofilotoxina , Tubulina (Proteína) , Humanos , Podofilotoxina/análogos & derivados , Podofilotoxina/farmacología , Podofilotoxina/química , Tubulina (Proteína)/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Supervivencia Celular/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Colorantes Fluorescentes/química , Sitios de Unión , Estrés del Retículo Endoplásmico/efectos de los fármacos
13.
J Appl Biomed ; 22(2): 99-106, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38912865

RESUMEN

Resveratrol (RSV) is a polyphenol antioxidant that has been shown to have neuroprotective effects. We sought molecular mechanisms that emphasize the anti-inflammatory activity of RSV in traumatic brain injury (TBI) in mice associated with endoplasmic reticulum stress (ERS). After establishing three experimental groups (sham, TBI, and TBI+RSV), we explored the results of RSV after TBI on ERS and caspase-12 apoptotic pathways. The expression levels of C/EBP homologous protein (CHOP), glucose regulated protein 78kD (GRP78), caspase-3, and caspase-12 in cortical brain tissues were assessed by western blotting. The qPCR analysis was also performed on mRNA expression of tumor necrosis factor (TNF)-α and interleukin (IL)-1ß in cortical brain tissue. In addition, the expression of GRP78 in microglia (ionized calcium binding adaptor molecule 1; Iba-1) and neurons (neuronal nuclei; NeuN) was identified by immunofluorescence staining. The neurological function of mice was assessed by modified neurological severity scores (mNSS). After drug treatment, the expression of CHOP, GRP78, caspase-3 and caspase-12 decreased, and qPCR results showed that TNF-α and IL-1ß were down-regulated. Immunofluorescence staining showed down-regulation of Iba-1+/GRP78+ and NeuN+/GRP78+ cells after RSV treatment. The mNSS analysis confirmed improvement after RSV treatment. RSV improved apoptosis by downregulating the ERS signaling pathway and improved neurological prognosis in mice with TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Resveratrol , Animales , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/metabolismo , Resveratrol/farmacología , Resveratrol/uso terapéutico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ratones , Masculino , Apoptosis/efectos de los fármacos , Pronóstico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Caspasa 12/metabolismo , Caspasa 12/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Factor de Necrosis Tumoral alfa/metabolismo , Ratones Endogámicos C57BL , Muerte Celular/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética
14.
Pharmacol Res ; 205: 107263, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38876442

RESUMEN

Pressure overload-induced pathological cardiac hypertrophy eventually leads to heart failure (HF). Unfortunately, lack of effective targeted therapies for HF remains a challenge in clinical management. Mixed-lineage leukemia 4 (MLL4) is a member of the SET family of histone methyltransferase enzymes, which possesses histone H3 lysine 4 (H3K4)-specific methyltransferase activity. However, whether and how MLL4 regulates cardiac function is not reported in adult HF. Here we report that MLL4 is required for endoplasmic reticulum (ER) stress homeostasis of cardiomyocytes and protective against pressure overload-induced cardiac hypertrophy and HF. We observed that MLL4 is increased in the heart tissue of HF mouse model and HF patients. The cardiomyocyte-specific deletion of Mll4 (Mll4-cKO) in mice leads to aggravated ER stress and cardiac dysfunction following pressure overloading. MLL4 knockdown neonatal rat cardiomyocytes (NRCMs) also display accelerated decompensated ER stress and hypertrophy induced by phenylephrine (PE). The combined analysis of Cleavage Under Targets and Tagmentation sequencing (CUT&Tag-seq) and RNA sequencing (RNA-seq) data reveals that, silencing of Mll4 alters the chromatin landscape for H3K4me1 modification and gene expression patterns in NRCMs. Interestingly, the deficiency of MLL4 results in a marked reduction of H3K4me1 and H3K27ac occupations on Thrombospondin-4 (Thbs4) gene loci, as well as Thbs4 gene expression. Mechanistically, MLL4 acts as a transcriptional activator of Thbs4 through mono-methylation of H3K4 and further regulates THBS4-dependent ER stress response, ultimately plays a role in HF. Our study indicates that pharmacologically targeting MLL4 and ER stress might be a valid therapeutic approach to protect against cardiac hypertrophy and HF.


Asunto(s)
Estrés del Retículo Endoplásmico , Insuficiencia Cardíaca , N-Metiltransferasa de Histona-Lisina , Ratones Endogámicos C57BL , Miocitos Cardíacos , Animales , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/etiología , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Masculino , Humanos , Ratones Noqueados , Ratas , Ratones , Células Cultivadas , Cardiomegalia/metabolismo , Cardiomegalia/genética , Ratas Sprague-Dawley , Trombospondinas
15.
Gut Microbes ; 16(1): 2367342, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38889450

RESUMEN

Alcohol-related liver disease (ALD) is recognized as a global health crisis, contributing to approximately 20% of liver cancer-associated fatalities. Dysbiosis of the gut microbiome is associated with the development of ALD, with the gut microbial metabolite urolithin A (UA) exhibiting a potential for alleviating liver symptoms. However, the protective efficacy of UA against ALD and its underlying mechanism mediated by microbiota remain elusive. In this study, we provide evidence demonstrating that UA effectively ameliorates alcohol-induced metabolic disorders and hepatic endoplasmic reticulum (ER) stress through a specific gut-microbiota-liver axis mediated by major urinary protein 1 (MUP1). Moreover, UA exhibited the potential to restore alcohol-induced dysbiosis of the intestinal microbiota by enriching the abundance of Bacteroides sartorii (B. sartorii), Parabacteroides distasonis (P. distasonis), and Akkermansia muciniphila (A. muciniphila), along with their derived metabolite propionic acid. Partial attenuation of the hepatoprotective effects exerted by UA was observed upon depletion of gut microbiota using antibiotics. Subsequently, a fecal microbiota transplantation (FMT) experiment was conducted to evaluate the microbiota-dependent effects of UA in ALD. FMT derived from mice treated with UA exhibited comparable efficacy to direct UA treatment, as it effectively attenuated ER stress through modulation of MUP1. It was noteworthy that strong associations were observed among the hepatic MUP1, gut microbiome, and metabolome profiles affected by UA. Intriguingly, oral administration of UA-enriched B. sartorii, P. distasonis, and A. muciniphila can enhance propionic acid production to effectively suppress ER stress via MUP1, mimicking UA treatment. Collectively, these findings elucidate the causal mechanism that UA alleviated ALD through the gut-microbiota-liver axis. This unique mechanism sheds light on developing novel microbiome-targeted therapeutic strategies against ALD.


Asunto(s)
Cumarinas , Estrés del Retículo Endoplásmico , Microbioma Gastrointestinal , Hepatopatías Alcohólicas , Hígado , Ratones Endogámicos C57BL , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Ratones , Hígado/metabolismo , Hígado/efectos de los fármacos , Hepatopatías Alcohólicas/microbiología , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/prevención & control , Masculino , Estrés del Retículo Endoplásmico/efectos de los fármacos , Cumarinas/farmacología , Cumarinas/metabolismo , Disbiosis/microbiología , Humanos , Bacterias/metabolismo , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación
16.
Biochem Biophys Res Commun ; 724: 150226, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38865815

RESUMEN

In patients with high-level radiation exposure, gastrointestinal injury is the main cause of death. Despite the severity of damage to the gastrointestinal tract, no specific therapeutic option is available. Tauroursodeoxycholic acid (TUDCA) is a conjugated form of ursodeoxycholic acid that suppresses endoplasmic reticulum (ER) stress and regulates various cell-signaling pathways. We investigated the effect of TUDCA premedication in alleviating intestinal damage and enhancing the survival of C57BL/6 mice administered a lethal dose (15Gy) of focal abdominal irradiation. TUDCA was administered to mice 1 h before radiation exposure, and reduced apoptosis of the jejunal crypts 12 h after irradiation. At later timepoint (3.5 days), irradiated mice manifested intestinal morphological changes that were detected via histological examination. TUDCA decreased the inflammatory cytokine levels and attenuated the decrease in serum citrulline levels after radiation exposure. Although radiation induced ER stress, TUDCA pretreatment decreased ER stress in the irradiated intestinal cells. The effect of TUDCA indicates the possibility of radiation therapy for cancer in tumor cells. TUDCA did not affect cell proliferation and apoptosis in the intestinal epithelium. TUDCA decreased the invasive ability of the CT26 metastatic colon cancer cell line. Reduced invasion after TUDCA treatment was associated with decreased matrix metalloproteinase (MMP)-7 and MMP-13 expression, which play important roles in invasion and metastasis. This study shows a potential role of TUDCA in protecting against radiation-induced intestinal damage and inhibiting tumor cell migration without any radiation and radiation therapy effect.


Asunto(s)
Apoptosis , Estrés del Retículo Endoplásmico , Ratones Endogámicos C57BL , Protectores contra Radiación , Ácido Tauroquenodesoxicólico , Animales , Ácido Tauroquenodesoxicólico/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de la radiación , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Protectores contra Radiación/farmacología , Ratones , Masculino , Intestinos/efectos de la radiación , Intestinos/efectos de los fármacos , Intestinos/patología , Modelos Animales de Enfermedad , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de la radiación , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Traumatismos Experimentales por Radiación/prevención & control , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación
17.
Sci Rep ; 14(1): 14004, 2024 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890434

RESUMEN

Cisplatin is an effective chemotherapeutic agent widely used for the treatment of various solid tumors. However, cisplatin has an important limitation in its use; currently, there is no method to ameliorate cisplatin-induced acute kidney injury (AKI). Thrombomodulin (TM) is well known not only for its role as a cofactor in the clinically important natural anticoagulation pathway but also for its anti-inflammatory properties. Here, we investigated the effects of TM in cisplatin-induced AKI. In mice intraperitoneally injected with 15 mg/kg cisplatin, TM (10 mg/kg) or PBS was administered intravenously at 24 h after cisplatin injection. TM significantly attenuated cisplatin-induced nephrotoxicity with the suppressed elevation of blood urea nitrogen and serum creatinine, and reduced histological damages. Actually, TM treatment significantly alleviated oxidative stress-induced apoptosis by reducing reactive oxygen species (ROS) levels in cisplatin-treated renal proximal tubular epithelial cells (RPTECs) in vitro. Furthermore, TM clarified cisplatin-induced apoptosis by reducing caspase-3 levels. In addition, TM attenuated the endoplasmic reticulum (ER) stress signaling pathway in both renal tissues and RPTECs to protect the kidneys from cisplatin-induced AKI. These findings suggest that TM is a potential protectant against cisplatin-induced nephrotoxicity through suppressing ROS generation and ER stress in response to cisplatin.


Asunto(s)
Lesión Renal Aguda , Apoptosis , Cisplatino , Estrés del Retículo Endoplásmico , Estrés Oxidativo , Especies Reactivas de Oxígeno , Trombomodulina , Cisplatino/efectos adversos , Animales , Trombomodulina/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/patología , Ratones , Especies Reactivas de Oxígeno/metabolismo , Masculino , Apoptosis/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Antineoplásicos/efectos adversos , Antineoplásicos/toxicidad , Ratones Endogámicos C57BL , Nitrógeno de la Urea Sanguínea , Transducción de Señal/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología
18.
Int J Oncol ; 65(2)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38873997

RESUMEN

Non­small cell lung cancer (NSCLC) is one of the major causes of cancer­related death worldwide. Cisplatin is a front­line chemotherapeutic agent in NSCLC. Nevertheless, subsequent harsh side effects and drug resistance limit its further clinical application. Polydatin (PD) induces apoptosis in various cancer cells by generating reactive oxygen species (ROS). However, underlying molecular mechanisms of PD and its effects on cisplatin­mediated antitumor activity in NSCLC remains unknown. MTT, colony formation, wound healing analyses and flow cytometry was employed to investigate the cell phenotypic changes and ROS generation. Relative gene and protein expressions were evaluated by reverse transcription­quantitative PCR and western blot analyses. The antitumor effects of PD, cisplatin and their combination were evaluated by mouse xenograft model. In the present study, it was found that PD in combination with cisplatin synergistically enhances the antitumor activity in NSCLC by stimulating ROS­mediated endoplasmic reticulum stress, and the C­Jun­amino­terminal kinase and p38 mitogen­activated protein kinase signaling pathways. PD treatment elevated ROS generation by promoting expression of NADPH oxidase 5 (NOX5), and NOX5 knockdown attenuated ROS­mediated cytotoxicity of PD in NSCLC cells. Mice xenograft model further confirmed the synergistic antitumor efficacy of combined therapy with PD and cisplatin. The present study exhibited a superior therapeutic strategy for some patients with NSCLC by combining PD and cisplatin.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Cisplatino , Sinergismo Farmacológico , Glucósidos , Neoplasias Pulmonares , NADPH Oxidasa 5 , Estrés Oxidativo , Especies Reactivas de Oxígeno , Estilbenos , Ensayos Antitumor por Modelo de Xenoinjerto , Cisplatino/farmacología , Cisplatino/uso terapéutico , Glucósidos/farmacología , Glucósidos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Animales , Humanos , Estilbenos/farmacología , Estilbenos/uso terapéutico , Ratones , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Apoptosis/efectos de los fármacos , Células A549 , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proliferación Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Masculino
19.
Neurotox Res ; 42(4): 31, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38935306

RESUMEN

Endoplasmic reticulum (ER) stress and oxidative stress (OS) are often related states in pathological conditions including Parkinson's disease (PD). This study investigates the role of anti-oxidant protein paraoxonase 2 (PON2) in ER stress and OS in PD, along with its regulatory molecule. PD was induced in C57BL/6 mice using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) treatment and in SH-SY5Y cells using 1-methyl-4-phenylpyridinium. PON2 was found to be poorly expressed in the substantia nigra pars compacta (SNc) of PD mice, and its overexpression improved motor coordination of mice. Through the evaluation of tyrosine hydroxylase, dopamine transporter, reactive oxygen species (ROS), and C/EBP homologous protein (CHOP) levels and neuronal loss in mice, as well as the examination of CHOP, glucose-regulated protein 94 (GRP94), GRP78, caspase-12, sarco/endoplasmic reticulum calcium ATPase 2, malondialdehyde, and superoxide dismutase levels in SH-SY5Y cells, we observed that PON2 overexpression mitigated ER stress, OS, and neuronal apoptosis both in vivo and in vitro. Forkhead box A1 (FOXA1) was identified as a transcription factor binding to the PON2 promoter to activate its transcription. Upregulation of FOXA1 similarly protected against neuronal loss by alleviating ER stress and OS, while the protective roles were abrogated by additional PON2 silencing. In conclusion, this study demonstrates that FOXA1-mediated transcription of PON2 alleviates ER stress and OS, ultimately reducing neuronal apoptosis in PD.


Asunto(s)
Apoptosis , Arildialquilfosfatasa , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Factor Nuclear 3-alfa del Hepatocito , Ratones Endogámicos C57BL , Estrés Oxidativo , Animales , Estrés del Retículo Endoplásmico/fisiología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Arildialquilfosfatasa/metabolismo , Arildialquilfosfatasa/genética , Humanos , Línea Celular Tumoral , Masculino , Ratones , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Factor Nuclear 3-alfa del Hepatocito/genética , Neuronas/metabolismo , Neuronas/efectos de los fármacos
20.
Mol Med Rep ; 30(2)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38904207

RESUMEN

Montelukast and zafirlukast, cysteinyl leukotriene receptor antagonists (LTRAs), trigger apoptosis and inhibit cell proliferation of triple­negative breast cancer MDA­MB­231 cells. By contrast, only zafirlukast induces G0/G1 cell cycle arrest. The present study compared the effects of these drugs on proteins regulating cell proliferation, apoptosis, autophagy, and endoplasmic reticulum (ER) and oxidative stress using reverse transcription­quantitative PCR, western blotting and flow cytometry. The expression of proliferating markers, Ki­67 and proliferating cell nuclear antigen, was decreased by both drugs. Zafirlukast, but not montelukast, decreased the expression of cyclin D1 and CDK4, disrupting progression from G1 to S phase. Zafirlukast also increased the expression of p27, a cell cycle inhibitor. Both drugs decreased the expression of anti­apoptotic protein Bcl­2 and ERK1/2 phosphorylation, and increased levels of the autophagy marker LC3­II and DNA damage markers, including cleaved PARP­1, phosphorylated (p)­ATM and p­histone H2AX. The number of caspase 3/7­positive cells was greater in montelukast­treated cells compared with zafirlukast­treated cells. Montelukast induced higher levels of the ER stress marker CHOP compared with zafirlukast. Montelukast activated PERK, activating transcription factor 6 (ATF6) and inositol­requiring enzyme type 1 (IRE1) pathways, while zafirlukast only stimulated ATF6 and IRE1 pathways. GSK2606414, a PERK inhibitor, decreased apoptosis mediated by montelukast, but did not affect zafirlukast­induced cell death. The knockdown of CHOP by small interfering RNA reduced apoptosis triggered by montelukast and zafirlukast. In conclusion, the effects on cell cycle regulator proteins may contribute to cell cycle arrest caused by zafirlukast. The greater apoptotic effects of montelukast may be caused by the higher levels of activated caspase enzymes and the activation of three pathways of ER stress: PERK, ATF6, and IRE1.


Asunto(s)
Acetatos , Apoptosis , Autofagia , Ciclopropanos , Daño del ADN , Estrés del Retículo Endoplásmico , Indoles , Quinolinas , Sulfuros , Sulfonamidas , Humanos , Sulfuros/farmacología , Ciclopropanos/farmacología , Quinolinas/farmacología , Apoptosis/efectos de los fármacos , Acetatos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Línea Celular Tumoral , Autofagia/efectos de los fármacos , Sulfonamidas/farmacología , Indoles/farmacología , Femenino , Daño del ADN/efectos de los fármacos , Fenilcarbamatos/farmacología , Compuestos de Tosilo/farmacología , Proliferación Celular/efectos de los fármacos , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Ciclo Celular/efectos de los fármacos , Antagonistas de Leucotrieno/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA