Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Inflamm Res ; 73(4): 597-617, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38353723

RESUMEN

OBJECTIVE: PANoptosis, a new form of regulated cell death, concomitantly manifests hallmarks for pyroptosis, apoptosis, and necroptosis. It has been usually observed in macrophages, a class of widely distributed innate immune cells in various tissues, upon pathogenic infections. The second-generation curaxin, CBL0137, can trigger necroptosis and apoptosis in cancer-associated fibroblasts. This study aimed to explore whether CBL0137 induces PANoptosis in macrophages in vitro and in mouse tissues in vivo. METHODS: Bone marrow-derived macrophages and J774A.1 cells were treated with CBL0137 or its combination with LPS for indicated time periods. Cell death was assayed by propidium iodide staining and immunoblotting. Immunofluorescence microscopy was used to detect cellular protein distribution. Mice were administered with CBL0137 plus LPS and their serum and tissues were collected for biochemical and histopathological analyses, respectively. RESULTS: The results showed that CBL0137 alone or in combination with LPS induced time- and dose-dependent cell death in macrophages, which was inhibited by a combination of multiple forms of cell death inhibitors but not each alone. This cell death was independent of NLRP3 expression. CBL0137 or CBL0137 + LPS-induced cell death was characterized by simultaneously increased hallmarks for pyroptosis, apoptosis and necroptosis, indicating that this is PANoptosis. Induction of PANoptosis was associated with Z-DNA formation in the nucleus and likely assembly of PANoptosome. ZBP1 was critical in mediating CBL0137 + LPS-induced cell death likely by sensing Z-DNA. Moreover, intraperitoneal administration of CBL0137 plus LPS induced systemic inflammatory responses and caused multi-organ (including the liver, kidney and lung) injury in mice due to induction of PANoptosis in these organs. CONCLUSIONS: CBL0137 alone or plus inflammatory stimulation induces PANoptosis both in vitro and in vivo, which is associated with systemic inflammatory responses in mice.


Asunto(s)
Carbazoles , ADN de Forma Z , Neoplasias , Ratones , Animales , Lipopolisacáridos/farmacología , Apoptosis , Piroptosis
2.
Acta Pharmacol Sin ; 45(3): 594-608, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37964019

RESUMEN

PANoptosis is a new type of cell death featured with pyroptosis, apoptosis and necroptosis, and is implicated in organ injury and mortality in various inflammatory diseases, such as sepsis and hemophagocytic lymphohistiocytosis (HLH). Reverse electron transport (RET)-mediated mitochondrial reactive oxygen species (mtROS) has been shown to contribute to pyroptosis and necroptosis. In this study we investigated the roles of mtROS and RET in PANoptosis induced by TGF-ß-activated kinase 1 (TAK1) inhibitor 5Z-7-oxozeaenol (Oxo) plus lipopolysaccharide (LPS) as well as the effects of anti-RET reagents on PANoptosis. We showed that pretreatment with anti-RET reagents 1-methoxy PMS (MPMS) or dimethyl fumarate (DMF) dose-dependently inhibited PANoptosis in macrophages BMDMs and J774A.1 cells induced by Oxo/LPS treatment assayed by propidium iodide (PI) staining. The three arms of the PANoptosis signaling pathway, namely pyroptosis, apoptosis and necroptosis signaling, as well as the formation of PANoptosomes were all inhibited by MPMS or DMF. We demonstrated that Oxo/LPS treatment induced RET and mtROS in BMDMs, which were reversed by MPMS or DMF pretreatment. Interestingly, the PANoptosome was co-located with mitochondria, in which the mitochondrial DNA was oxidized. MPMS and DMF fully blocked the mtROS production and the formation of PANoptosome induced by Oxo plus LPS treatment. An HLH mouse model was established by poly(I:C)/LPS challenge. Pretreatment with DMF (50 mg·kg-1·d-1, i.g. for 3 days) or MPMS (10 mg·kg-1·d-1, i.p. for 2 days) (DMF i.g. MPMS i.p.) effectively alleviated HLH lesions accompanied by decreased hallmarks of PANoptosis in the liver and kidney. Collectively, RET and mtDNA play crucial roles in PANoptosis induction and anti-RET reagents represent a novel class of PANoptosis inhibitors by blocking oxidation of mtDNA, highlighting their potential application in treating PANoptosis-related inflammatory diseases. PANoptotic stimulation induces reverse electron transport (RET) and reactive oxygen species (ROS) in mitochondia, while 1-methoxy PMS and dimethyl fumarate can inhibit PANoptosis by suppressing RETmediated oxidation of mitochondrial DNA.


Asunto(s)
ADN Mitocondrial , Dimetilfumarato , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Transporte de Electrón , Dimetilfumarato/metabolismo , Dimetilfumarato/farmacología , ADN Mitocondrial/metabolismo , Lipopolisacáridos/farmacología , Electrones , Mitocondrias , Apoptosis
3.
Apoptosis ; 28(11-12): 1646-1665, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37702860

RESUMEN

Macrophages represent the first lines of innate defense against pathogenic infections and are poised to undergo multiple forms of regulated cell death (RCD) upon infections or toxic stimuli, leading to multiple organ injury. Triptolide, an active compound isolated from Tripterygium wilfordii Hook F., possesses various pharmacological activities including anti-tumor and anti-inflammatory effects, but its applications have been hampered by toxic adverse effects. It remains unknown whether and how triptolide induces different forms of RCD in macrophages. In this study, we showed that triptolide exhibited significant cytotoxicity on cultured macrophages in vitro, which was associated with multiple forms of lytic cell death that could not be fully suppressed by any one specific inhibitor for a single form of RCD. Consistently, triptolide induced the simultaneous activation of pyroptotic, apoptotic and necroptotic hallmarks, which was accompanied by the co-localization of ASC specks respectively with RIPK3 or caspase-8 as well as their interaction with each other, indicating the formation of PANoptosome and thus the induction of PANoptosis. Triptolide-induced PANoptosis was associated with mitochondrial dysfunction and ROS production. PANoptosis was also induced by triptolide in mouse peritoneal macrophages in vivo. Furthermore, triptolide caused kidney and liver injury, which was associated with systemic inflammatory responses and the activation of hallmarks for PANoptosis in vivo. Collectively, our data reveal that triptolide induces PANoptosis in macrophages in vitro and exhibits nephrotoxicity and hepatotoxicity associated with induction of PANoptosis in vivo, suggesting a new avenue to alleviate triptolide's toxicity by harnessing PANoptosis.


Asunto(s)
Diterpenos , Fenantrenos , Ratones , Animales , Apoptosis , Macrófagos/metabolismo , Diterpenos/efectos adversos , Diterpenos/metabolismo , Fenantrenos/toxicidad , Fenantrenos/metabolismo , Compuestos Epoxi/toxicidad , Compuestos Epoxi/metabolismo
4.
Acta Pharmacol Sin ; 44(10): 2019-2036, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37221235

RESUMEN

Activation of NLR family pyrin domain-containing 3 (NLRP3) inflammasome plays important role in defending against infections, but its aberrant activation is causally linked to many inflammatory diseases, thus being a therapeutic target for these diseases. Theaflavin, one major ingredient of black tea, exhibits potent anti-inflammatory and anti-oxidative activities. In this study, we investigated the therapeutic effects of theaflavin against NLRP3 inflammasome activation in macrophages in vitro and in animal models of related diseases. We showed that theaflavin (50, 100, 200 µM) dose-dependently inhibited NLRP3 inflammasome activation in LPS-primed macrophages stimulated with ATP, nigericin or monosodium urate crystals (MSU), evidenced by reduced release of caspase-1p10 and mature interleukin-1ß (IL-1ß). Theaflavin treatment also inhibited pyroptosis as shown by decreased generation of N-terminal fragment of gasdermin D (GSDMD-NT) and propidium iodide incorporation. Consistent with these, theaflavin treatment suppressed ASC speck formation and oligomerization in macrophages stimulated with ATP or nigericin, suggesting reduced inflammasome assembly. We revealed that theaflavin-induced inhibition on NLRP3 inflammasome assembly and pyroptosis resulted from ameliorated mitochondrial dysfunction and reduced mitochondrial ROS production, thereby suppressing interaction between NLRP3 and NEK7 downstream of ROS. Moreover, we showed that oral administration of theaflavin significantly attenuated MSU-induced mouse peritonitis and improved the survival of mice with bacterial sepsis. Consistently, theaflavin administration significantly reduced serum levels of inflammatory cytokines including IL-1ß and attenuated liver inflammation and renal injury of mice with sepsis, concomitant with reduced generation of caspase-1p10 and GSDMD-NT in the liver and kidney. Together, we demonstrate that theaflavin suppresses NLRP3 inflammasome activation and pyroptosis by protecting mitochondrial function, thus mitigating acute gouty peritonitis and bacterial sepsis in mice, highlighting a potential application in treating NLRP3 inflammasome-related diseases.


Asunto(s)
Gota , Peritonitis , Sepsis , Ratones , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Especies Reactivas de Oxígeno , Nigericina/uso terapéutico , Peritonitis/tratamiento farmacológico , Antioxidantes/uso terapéutico , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , Caspasas , Adenosina Trifosfato , Interleucina-1beta/metabolismo
5.
Acta Pharmacol Sin ; 43(5): 1324-1336, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-34376811

RESUMEN

Monosodium urate (MSU) crystals, the etiological agent of gout, are formed in joints and periarticular tissues due to long-lasting hyperuricemia. Although MSU crystal-triggered NLRP3 inflammasome activation and interleukin 1ß (IL-1ß) release are known to have key roles in gouty arthritis, recent studies revealed that MSU crystal-induced necrosis also plays a critical role in this process. However, it remains unknown what forms of necrosis have been induced and whether combined cell death inhibitors can block such necrosis. Here, we showed that MSU crystal-induced necrosis in murine macrophages was not dependent on NLRP3 inflammasome activation, as neither genetic deletion nor pharmacological blockade of the NLRP3 pathway inhibited the necrosis. Although many cell death pathways (such as ferroptosis and pyroptosis) inhibitors or reactive oxygen species inhibitors did not have any suppressive effects, necroptosis pathway inhibitors GSK'872 (RIPK3 inhibitor), and GW806742X (MLKL inhibitor) dose-dependently inhibited MSU crystal-induced necrosis. Moreover, a triple combination of GSK'872, GW806742X, and IDN-6556 (pan-caspase inhibitor) displayed enhanced inhibition of the necrosis, which was further fortified by the addition of MCC950 (NLRP3 inhibitor), suggesting that multiple cell death pathways might have been triggered by MSU crystals. Baicalin, a previously identified inhibitor of NLRP3, inhibited MSU crystal-induced inflammasome activation and suppressed the necrosis in macrophages. Besides, baicalin gavage significantly ameliorated MSU crystal-induced peritonitis in mice. Altogether, our data indicate that MSU crystals induce NLRP3-independent necrosis, which can be inhibited by combined inhibitors for multiple signaling pathways, highlighting a new avenue for the treatment of gouty arthritis.


Asunto(s)
Artritis Gotosa , Gota , Animales , Artritis Gotosa/inducido químicamente , Artritis Gotosa/tratamiento farmacológico , Artritis Gotosa/metabolismo , Gota/tratamiento farmacológico , Gota/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Necrosis/inducido químicamente , Necrosis/tratamiento farmacológico , Transducción de Señal , Ácido Úrico
6.
Int Ophthalmol ; 41(3): 845-852, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33174070

RESUMEN

OBJECTIVE: To observe the effect of sirt1 on retinal ganglion cells (RGC) with high glucose culture and to explore the role of sirt1 in the development of diabetic retinopathy. Method RGC was infected by sirt1 lentivirus overexpression vector pLV5-sirt1 and interference vector pLV3-si-sirt1. The normal control group and control virus vector group were set up at the same time. After 48 h of infection, the viability of RGC was detected by CCK8 kit, the apoptosis rate was detected by FCM analysis, and the protein expression of p53, FOXO3a, NF-κ B, caspase-3 was detected by Western blot. RESULTS: After RGC were infected with lentivirus, the cell viability of lentivirus overexpression vector pLV5-sirt1 was significantly higher than that of the high glucose group and the sirt1 overexpression control group, while the cell viability of interference vector pLV3-si-sirt1 was significantly lower than that of the high glucose group and the sirt1 interference control group (P < 0.05). At the same time, the apoptosis rate of RGC cells infected by lentivirus overexpression vector pLV5-sirt1 was lower than that of the high glucose group and the control virus vector group, while the apoptosis rate of the interference vector pLV3-si-sirt1 cells was significantly higher than that of the high glucose group and the control virus vector group (P < 0.05). The results of Western blotting showed that the expression of p53, FOXO3a, NF-κ B and caspase-3 in RGC cells decreased significantly after infection with pLV5-sirt1 compared with the high glucose group and the control virus vector group, while the expression of p53, FOXO3a, NF-κB and caspase-3 in RGC cells increased significantly after infection with pLV3-si-sirt1 (P < 0.05). CONCLUSION: Sirt1 can inhibit the apoptosis of RGCs through regulating the expression of some apoptotic cytokinessome, and it can be used as a candidate gene for the biotherapy of retinal diseases.


Asunto(s)
Células Ganglionares de la Retina , Sirtuina 1 , Animales , Apoptosis , Línea Celular , Supervivencia Celular , Glucosa , Ratones , Sirtuina 1/genética
7.
Apoptosis ; 24(3-4): 312-325, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30710195

RESUMEN

Gasdermin E (GSDME) has an important role in inducing secondary necrosis/pyroptosis. Upon apoptotic stimulation, it can be cleaved by activated caspase-3 to generate its N-terminal fragment (GSDME-NT), which executes pyroptosis by perforating the plasma membrane. GSDME is expressed in many human lung cancers including A549 cells. Paclitaxel and cisplatin are two representative chemotherapeutic agents for lung cancers, which induce apoptosis via different action mechanisms. However, it remains unclear whether they can induce GSDME-mediated secondary necrosis/pyroptosis in lung A549 cancer cells. Here we showed that both paclitaxel and cisplatin evidently induced apoptosis in A549 cells as revealed by the activation of multiple apoptotic markers. Notably, some of the dying cells displayed characteristic morphology of secondary necrosis/pyroptosis, by blowing large bubbles from the cellular membrane accompanied by caspase-3 activation and GSDME-NT generation. But the ability of cisplatin to induce this phenomenon was much stronger than that of paclitaxel. Consistent with this, cisplatin triggered much higher activation of caspase-3 and generation of GSDME-NT than paclitaxel, suggesting that the levels of secondary necrosis/pyroptosis correlated with the levels of active caspase-3 and GSDME-NT. Supporting this, caspase-3 specific inhibitor (Ac-DEVD-CHO) suppressed cisplatin-induced GSDME-NT generation and concurrently reduced the secondary necrosis/pyroptosis. Besides, GSDME knockdown significantly inhibited cisplatin- but not paclitaxel-induced secondary necrosis/pyroptosis. These results indicated that cisplatin induced higher levels of secondary necrosis/pyroptosis in A549 cells than paclitaxel, suggesting that cisplatin may provide additional advantages in the treatment of lung cancers with high levels of GSDME expression.


Asunto(s)
Antineoplásicos/farmacología , Caspasa 3/metabolismo , Cisplatino/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Paclitaxel/farmacología , Piroptosis/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Células A549 , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/metabolismo , Necrosis/tratamiento farmacológico , Necrosis/metabolismo
8.
Apoptosis ; 24(9-10): 703-717, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31175486

RESUMEN

ATP acts as a canonical activator to induce NLRP3 (NOD-like receptor family, pyrin domain containing 3) inflammasome activation in macrophages, leading to caspase-1/gasdermin D (GSDMD)-mediated pyroptosis. It remains unclear whether ATP can induce pyroptosis in macrophages when the NLRP3 pathway is blocked by pathogenic infection. In this study, we used cellular models to mimic such blockade of NLRP3 activation: bone marrow-derived macrophages (BMDMs) treated with NLRP3-specific inhibitor MCC950 and RAW264.7 cells deficient in ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain) expression. The results showed that ATP treatment induced lytic cell death morphologically resembling canonical pyroptosis in both MCC950-treated BMDMs and RAW264.7 cells, but did not cause the activation of caspase-1 (by detecting caspase-1p10 and mature interleukin-1ß) and cleavage of GSDMD. Instead, both apoptotic initiator (caspase-8 and -9) and executioner (caspase-3 and -7) caspases were evidently activated and gasdermin E (GSDME) was cleaved to generate its N-terminal fragment (GSDME-NT) which executes pyroptosis. The GSDME-NT production and lytic cell death induced by ATP were diminished by caspase-3 inhibitor. In BMDMs without MCC950 treatment, ATP induced the formation of ASC specks which were co-localized with caspase-8; with MCC950 treatment, however, ATP did not induced the formation of ASC specks. In RAW264.7 cells, knockdown of GSDME by small interfering RNA attenuated ATP-induced lytic cell death and HMGB1 release into culture supernatants. Collectively, our results indicate that ATP induces pyroptosis in macrophages through the caspase-3/GSDME axis when the canonical NLRP3 pathway is blocked, suggestive of an alternative mechanism for combating against pathogen evasion.


Asunto(s)
Adenosina Trifosfato/farmacología , Caspasa 3/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Neoplasias/metabolismo , Piroptosis/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Caspasa 1/metabolismo , Caspasa 8/metabolismo , Caspasas/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Células RAW 264.7 , Interferencia de ARN
9.
Chemphyschem ; 18(12): 1611-1619, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28297133

RESUMEN

The abnormal fibrillation of human islet amyloid polypeptide (hIAPP) is associated with development of type II diabetes mellitus (T2DM). (-)-Epigallocatechin gallate (EGCG) can bind amyloid proteins to inhibit the fibrillation of these proteins. However, the mechanic detail of EGCG inhibiting amyloid formation is still unclear at the molecular level. In the present work, we sought to investigate the effect of EGCG on amidated hIAPP (hIAPP-NH2 ) fibrillation and aggregation by using spectroscopic and microscopic techniques, and also sought to gain insights into the interaction of EGCG and hIAPP22-27 by using spectroscopic experiments and quantum chemical calculations. ThT fluorescence, real-time NMR, and TEM studies demonstrated that EGCG inhibits the formation of hIAPP-NH2 fibrils, while promoting the formation of hIAPP-NH2 amorphous aggregates. Phenylalanine intrinsic fluorescence and NMR studies of the EGCG/hIAPP22-27 complex revealed three important binding sites including the A ring of EGCG, residue Phe23, and residue Ile26. DFT calculations identified the dominant binding structures of EGCG/Phe23 and EGCG/Ile26 complexes, named structure I and structure II, respectively. Our study demonstrates the inhibitory mechanism of EGCG on fibrillation and aggregation of hIAPP-NH2 in which EGCG interacts with hIAPP-NH2 through hydrogen bonding and π-π interactions between the A ring and residue Phe23 as well as hydrophobic interactions between the A ring and residue Ile26, which can thus inhibit the interpeptide interaction between hIAPP-NH2 monomers and finally inhibit fibrillation of hIAPP-NH2 . This study agrees with and reinforces previous studies and offers an intuitive explanation at both the atomic and molecular levels. Our findings may provide an invaluable reference for the future development of new drugs in the management of diabetes.


Asunto(s)
Catequina/análogos & derivados , Polipéptido Amiloide de los Islotes Pancreáticos/efectos de los fármacos , Catequina/farmacología , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química , Estructura Molecular , Agregado de Proteínas/efectos de los fármacos
10.
Apoptosis ; 21(2): 130-42, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26531131

RESUMEN

CPT-11 (Irinotecan) is a first-line chemotherapeutic agent in clinic, but it may induce side effects including diarrhea and enteritis in patients. The underlying mechanism of CPT-11's intestinal toxicity is unclear. Peritoneal resident macrophages have been reported to be important for the maintenance of intestinal homeostasis. In this study, we evaluated the cytotoxic effects of CPT-11 on mouse peritoneal resident macrophages. CPT-11 was administered intraperitoneally to mice and their peritoneal exudate cells were isolated for evaluation. CPT-11 treatment strikingly decreased the ratio of F4/80(hi)MHCII(low) large peritoneal macrophages (LPMs), which are regarded as prenatally-originated peritoneal resident macrophages. Consistent with this, the transcription factor GATA6 specifically expressed in LPMs was barely detectable in the macrophages from CPT-11-treated mice, indicative of elimination of LPMs. Such elimination of LPMs was at least partly due to CPT-induced apoptosis in macrophages, because inhibition of apoptosis by caspase-3 inhibitor z-DEVD-fmk significantly diminished the loss of GATA6(+) LPMs. As GATA6 is a transcription factor that controls expression of multiple genes regulating peritoneal B-1 cell development and translocation, elimination of GATA6(+) LPMs led to a great reduction in B-1 cells in the peritoneal cavity after CPT-11 treatment. These results indicated that CPT-11-induced apoptosis contributed to the elimination of peritoneal resident macrophages, which might in turn impair the function of peritoneal B-1 cells in maintaining intestinal homeostasis. Our findings may at least partly explain why CPT-11 treatment in cancer patients induces diarrhea and enteritis, which may provide a novel avenue to prevent such side effects.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Apoptosis/efectos de los fármacos , Camptotecina/análogos & derivados , Macrófagos Peritoneales/fisiología , Animales , Antineoplásicos Fitogénicos/efectos adversos , Camptotecina/administración & dosificación , Camptotecina/efectos adversos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Enteritis/inducido químicamente , Femenino , Inyecciones Intraperitoneales , Irinotecán , Macrófagos Peritoneales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7
11.
Toxicol Appl Pharmacol ; 292: 56-64, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26765310

RESUMEN

Gossypol, a polyphenolic compound isolated from cottonseeds, has been reported to possess many pharmacological activities, but whether it can influence inflammasome activation remains unclear. In this study, we found that in mouse macrophages, gossypol induced cell death characterized by rapid membrane rupture and robust release of HMGB1 and pro-caspase-11 comparable to ATP treatment, suggesting an induction of pyroptotic cell death. Unlike ATP, gossypol induced much low levels of mature interleukin-1ß (IL-1ß) secretion from mouse peritoneal macrophages primed with LPS, although it caused pro-IL-1ß release similar to that of ATP. Consistent with this, activated caspase-1 responsible for pro-IL-1ß maturation was undetectable in gossypol-treated peritoneal macrophages. Besides, RAW 264.7 cells lacking ASC expression and caspase-1 activation also underwent pyroptotic cell death upon gossypol treatment. In further support of pyroptosis induction, both pan-caspase inhibitor and caspase-1 subfamily inhibitor, but not caspase-3 inhibitor, could sharply suppress gossypol-induced cell death. Other canonical pyroptotic inhibitors, including potassium chloride and N-acetyl-l-cysteine, could suppress ATP-induced pyroptosis but failed to inhibit or even enhanced gossypol-induced cell death, whereas nonspecific pore-formation inhibitor glycine could attenuate this process, suggesting involvement of a non-canonical pathway. Of note, gossypol treatment eliminated thioglycollate-induced macrophages in the peritoneal cavity with recruitment of other leukocytes. Moreover, gossypol administration markedly decreased the survival of mice in a bacterial sepsis model. Collectively, these results suggested that gossypol induced pyroptosis in mouse macrophages via a non-canonical inflammasome pathway, which raises a concern for its in vivo cytotoxicity to macrophages.


Asunto(s)
Gosipol/toxicidad , Inflamasomas/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Piroptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular , Relación Dosis-Respuesta a Droga , Ratones , Ratones Endogámicos C57BL , Piroptosis/fisiología , Transducción de Señal/fisiología
12.
Poult Sci ; 95(3): 541-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26574032

RESUMEN

This study investigated the effects of xanthophylls (containing 40% lutein and 60% zeaxanthin; Juyuan Biochemical Co., Ltd., GuangZhou, China) on gene expression associated with carotenoid cleavage enzymes (ß-carotene 15, 15'-monooxygenase, BCMO1; and ß-carotene 9', 10'-dioxygenase, BCDO2) and retinoid metabolism (lecithin:retinol acyl transferase (LRAT) and STRA6) of breeding hens and chicks. In experiment 1, 432 hens were divided into 3 groups and fed diets supplemented with zero (as the control group), 20, or 40 mg/kg xanthophyll. The liver, duodenum, jejunum, and ileum were sampled at d 35 of the trial. Results showed that 40 mg/kg xanthophyll supplementation increased BCDO2 mRNA in the liver, duodenum, and jejunum; LRAT mRNA in the jejunum; and STRA6 mRNA in the liver, while it decreased LRAT mRNA in the liver. Experiment 2 was a 2 × 2 factorial design. Male chicks hatched from a zero or 40 mg/kg xanthophyll diet of hens were fed a diet containing either zero or 40 mg/kg xanthophylls. The liver, duodenum, jejunum, and ileum were sampled at zero, 7, 14, and 21 d after hatching. Results showed that in ovo xanthophyll modulated carotenoid and retinoid metabolism mainly within one wk after hatching. The maternal effects gradually vanished and dietary effects began to work one to 2 wk after hatching. Dietary xanthophyll regulated carotenoid and retinoid metabolism mainly from 2 wk onward. The xanthophyll regulation of carotenoid and retinoid metabolism also revealed strong tissue specificity. In conclusion, xanthophyll supplementation could modulate carotenoid and retinoid metabolism in different tissues of hens and chicks.


Asunto(s)
Carotenoides/metabolismo , Pollos/metabolismo , Dieta/veterinaria , Suplementos Dietéticos , Luteína/metabolismo , Retinoides/metabolismo , Zeaxantinas/metabolismo , Alimentación Animal/análisis , Animales , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Femenino , Regulación de la Expresión Génica , Intestino Delgado/metabolismo , Hígado/metabolismo , Masculino , Distribución Aleatoria
13.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 35(7): 778-82, 2015 Jul.
Artículo en Zh | MEDLINE | ID: mdl-26380437

RESUMEN

OBJECTIVE: To observe the protection of Qingyuan Shenghua Decoction (QSD) on multiple organs of sepsis patients after bone trauma, and to preliminarily explore its mechanism. METHODS: Totally 60 sepsis patients after bone trauma were randomly assigned to the treatment group and the control group according to random digit table, 30 in each group. All patients received routine Western medical treatment. Patients in the treatment group additionally took QSD or were nasally fed with QSD, one dose per day for 1 week. Changes of WBC, oxygenation index (PaO2/FiO2), serum creatinine (SCr), total bilirubin (TBIL), aspartate aminotransferase (AST), fibrinogen (FIB), D-dimer (DD), activated partial thromboplastin time (APTT), pro-calcitonin (PCT), C-reactive protein (CRP), heart rate (HR), mean arterial pressure (MAP), intra-abdominal pressure, scores for Acute Physiology and Chronic Health Evaluation II (APACHE II), sequential organ failure assessment (SOFA) scores were observed before treatment and on day 1, 3 and 7 after treatment. RESULTS: Compared with the control group at the same time point, MAP increased at post-treatment day 1 and 3; CRP, APTT, HR, SCr, TBIL, AST, intra-abdominal pressure at post-treatment day 3 obviously decreased in the treatment group (P < 0.05, P < 0.01). WBC, SOFA scores, PCT, CRP, APACHE II, APTT, D-D, HR, SCr, TBIL, AST and intra-abdominal pressure significantly decreased; FIB, MAP and PaO2/FiO2 obviously increased at post-treatment day 7 (P < 0.05, P < 0.01). CONCLUSION: QSD had good protective effect on multiple organ function in sepsis patients after bone trauma, and its mechanism might be related with effectively clearing endotoxin, alleviating inflammatory reactions, and fighting against coagulation dysfunction.


Asunto(s)
Enfermedades Óseas/complicaciones , Medicamentos Herbarios Chinos/farmacología , Sepsis/tratamiento farmacológico , APACHE , Coagulación Sanguínea , Proteína C-Reactiva/metabolismo , Calcitonina/metabolismo , Medicamentos Herbarios Chinos/uso terapéutico , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Humanos , Inflamación , Tiempo de Tromboplastina Parcial , Precursores de Proteínas/metabolismo , Sepsis/etiología
14.
Inflammation ; 47(1): 285-306, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37759136

RESUMEN

Itaconate is an unsaturated dicarboxylic acid that is derived from the decarboxylation of the Krebs cycle intermediate cis-aconitate and has been shown to exhibit anti-inflammatory and anti-bacterial/viral properties. But the mechanisms underlying itaconate's anti-inflammatory activities are not fully understood. Necroptosis, a lytic form of regulated cell death (RCD), is mediated by receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like protein (MLKL) signaling. It has been involved in the pathogenesis of organ injury in many inflammatory diseases. In this study, we aimed to explore whether itaconate and its derivatives can inhibit necroptosis in murine macrophages, a mouse MPC-5 cell line and a human HT-29 cell line in response to different necroptotic activators. Our results showed that itaconate and its derivatives dose-dependently inhibited necroptosis, among which dimethyl itaconate (DMI) was the most effective one. Mechanistically, itaconate and its derivatives inhibited necroptosis by suppressing the RIPK1/RIPK3/MLKL signaling and the oligomerization of MLKL. Furthermore, DMI promoted the nuclear translocation of Nrf2 that is a critical regulator of intracellular redox homeostasis, and reduced the levels of intracellular reactive oxygen species (ROS) and mitochondrial superoxide (mtROS) that were induced by necroptotic activators. Consistently, DMI prevented the loss of mitochondrial membrane potential induced by the necroptotic activators. In addition, DMI mitigated caerulein-induced acute pancreatitis in mice accompanied by reduced activation of the necroptotic signaling in vivo. Collectively, our study demonstrates that itaconate and its derivatives can inhibit necroptosis by suppressing the RIPK1/RIPK3/MLKL signaling, highlighting their potential applications for treating necroptosis-associated diseases.


Asunto(s)
Pancreatitis , Proteínas Quinasas , Succinatos , Ratones , Humanos , Animales , Proteínas Quinasas/metabolismo , Enfermedad Aguda , Antiinflamatorios , Apoptosis
15.
J Cell Biochem ; 114(10): 2415-29, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23695982

RESUMEN

Accumulating evidence indicates that cucurbitacin B (CuB), as well as other cucurbitacins, damages the actin cytoskeleton in a variety of cell types. However, the underlying mechanism of such an effect is not well understood. In this study, we showed that CuB rapidly induced actin aggregation followed by actin rod formation in melanoma cells. Cofilin, a critical regulator of actin dynamics, was dramatically dephosphorylated (i.e., activated) upon CuB treatment. Notably, the activated cofilin subsequently formed rod-like aggregates, which were highly colocalized with actin rods, indicating the formation of cofilin-actin rods. Cofilin knockdown significantly suppressed rod formation but did not prevent actin aggregation. Furthermore, knockdown of the cofilin phosphatase Slingshot homolog 1 (SSH1), but not chronophin (CIN), alleviated CuB-induced cofilin hyperactivation and cofilin-actin rod formation. The activity of Rho kinase and LIM kinase, two upstream regulators of cofilin activation, was downregulated after cofilin hyperactivation. Pretreatment with a thiol-containing reactive oxygen species (ROS) scavenger N-acetyl cysteine, but not other ROS inhibitors without thiol groups, suppressed CuB-induced actin aggregation, cofilin hyperactivation and cofilin-actin rod formation, suggesting that thiol oxidation might be involved in these processes. Taken together, our results demonstrated that CuB-induced formation of cofilin-actin rods was mediated by SSH1-dependent but CIN-independent cofilin hyperactivation.


Asunto(s)
Actinas/metabolismo , Cofilina 1/metabolismo , Cucurbitacinas/farmacología , Fosfoproteínas Fosfatasas/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Microscopía Fluorescente , Especies Reactivas de Oxígeno/metabolismo
16.
Biochem Biophys Res Commun ; 441(4): 970-5, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24220335

RESUMEN

Autophagy is a conserved mechanism for controlling the degradation of misfolded proteins and damaged organelles in eukaryotes and can be induced by nutrient withdrawal, including serum starvation. Although differential acetylation of autophagy-related proteins has been reported to be involved in autophagic flux, the regulation of acetylated microtubule-associated protein 1 light chain 3 (LC3) is incompletely understood. In this study, we found that the acetylation levels of phosphotidylethanolamine (PE)-conjugated LC3B (LC3B-II), which is a critical component of double-membrane autophagosome, were profoundly decreased in HeLa cells upon autophagy induction by serum starvation. Pretreatment with lysosomal inhibitor chloroquine did not attenuate such deacetylation. Under normal culture medium, we observed increased levels of acetylated LC3B-II in cells treated with tubacin, a specific inhibitor of histone deacetylase 6 (HDAC6). However, tubacin only partially suppressed serum-starvation-induced LC3B-II deacetylation, suggesting that HDAC6 is not the only deacetylase acting on LC3B-II during serum-starvation-induced autophagy. Interestingly, tubacin-induced increase in LC3B-II acetylation was associated with p62/SQSTM1 accumulation upon serum starvation. HDAC6 knockdown did not influence autophagosome formation but resulted in impaired degradation of p62/SQSTM1 during serum starvation. Collectively, our data indicated that LC3B-II deacetylation, which was partly mediated by HDAC6, is involved in autophagic degradation during serum starvation.


Asunto(s)
Autofagia/fisiología , Histona Desacetilasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Acetilación , Anilidas/farmacología , Autofagia/efectos de los fármacos , Medio de Cultivo Libre de Suero/farmacología , Técnicas de Silenciamiento del Gen , Células HeLa , Histona Desacetilasa 6 , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Humanos , Ácidos Hidroxámicos/farmacología , Lisosomas/fisiología , Fusión de Membrana , Proteolisis
17.
Inflamm Res ; 62(5): 461-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23360962

RESUMEN

OBJECTIVE: Cucurbitacin E (CuE), a triterpenoid compound isolated from Cucurbitaceae plants, possesses a wide range of biological activities including anti-inflammatory properties. The present study aimed to investigate the anti-inflammatory effect of CuE and the underlying mechanism of action. METHODS: The anti-inflammatory effect of CuE was evaluated in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. Cell proliferation was assessed using a modified MTT assay. Cell cycle distribution was analyzed by propidium iodide staining. The actin cytoskeleton was examined by immunofluorescent staining. The expression of tumor necrosis factor (TNF)-α and interleukin (IL)-1ß was determined by intracellular cytokine staining. G-actin level and nuclear factor (NF)-κB nuclear translocation were detected by immunoblotting. RESULTS: CuE inhibited cell proliferation and induced cell cycle arrest at G2/M phase in RAW 264.7 cells. CuE also suppressed LPS-induced cell spreading and pseudopodia formation. These effects were associated with decreased G-actin level and severe actin aggregation. Moreover, CuE significantly inhibited both TNF-α and IL-1ß production in LPS-stimulated RAW 264.7 cells. This was likely mediated by suppressing LPS-induced nuclear translocation of NF-κB, a critical transcription factor responsible for pro-inflammatory cytokine expression. CONCLUSION: CuE displayed anti-inflammatory effects through suppression of NF-κB nuclear translocation leading to a decreased expression of TNF-α and IL-1ß in LPS-stimulated RAW 264.7 cells.


Asunto(s)
Antiinflamatorios/farmacología , FN-kappa B/metabolismo , Triterpenos/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Interleucina-1beta/metabolismo , Lipopolisacáridos , Ratones , Factor de Necrosis Tumoral alfa/metabolismo
18.
Zhonghua Wai Ke Za Zhi ; 51(9): 827-30, 2013 Sep.
Artículo en Zh | MEDLINE | ID: mdl-24330965

RESUMEN

OBJECTIVE: To demonstrate the common characteristics of giant cell tumor of bone in immature skeletons. METHODS: From 1989 to 2009, the 8 skeletal immature patients were pathologically diagnosed with giant cell tumor (GCT) in our department, which accounted for 1.3% (8/621) of all GCT patients in an extremity. All patients were identified with an open epiphyseal plate by retrospective review of the radiograph, CT or MRI by senior consultants. Oncological and functional outcome were followed for a mean 44.1 months. There were 5 boys and 3 girls. The mean age was 13.8 years. All cases had a primary lesion. The distal femur is the most common site involved (3 cases), followed by the proximal tibia (2 cases). The proximal humerus, the distal tibia and the distal radius accounted for 1 case respectively. Oncological and functional outcome are followed for a mean 44.1 months. RESULTS: All lesions were lytic. Six lesions involved both the epiphysis and metaphysis. Two lesions located in the metaphysis area. Six lesions were treated with extended curettage and were reconstructed with allograft and (or) bone cement. Internal fixations were used in 2 cases. Two cases were treated with segmental resection. And one was reconstructed with cement spacer and the other one with segmental allograft and internal fixation. One patient (1/6) developed a bone recurrence after extended curettage. No extremity deformity and discrepancy were found during the follow up after the curettage. No metastasis was found during the follow up. CONCLUSION: Histologically GCT occurs in skeletal immature bone has the same pathological appearance but radiologically has its unique features. These lesions share same behavior as that in adults. A low local recurrence rate and good function can be achieved after a proper surgery.


Asunto(s)
Neoplasias Óseas , Tumor Óseo de Células Gigantes , Neoplasias Óseas/cirugía , Estudios de Seguimiento , Humanos , Recurrencia Local de Neoplasia , Estudios Retrospectivos
19.
Int Immunopharmacol ; 117: 109974, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37012867

RESUMEN

Necroptosis is a necrotic form of regulated cell death, which is primarily mediated by the receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like (MLKL) pathway in a caspase-independent manner. Necroptosis has been found to occur in virtually all tissues and diseases evaluated, including pancreatitis. Celastrol, a pentacyclic triterpene extracted from the roots of Tripterygium wilfordii (thunder god vine), possesses potent anti-inflammatory and anti-oxidative activities. Yet, it is unclear whether celastrol has any effects on necroptosis and necroptotic-related diseases. Here we showed that celastrol significantly suppressed necroptosis induced by lipopolysaccharide (LPS) plus pan-caspase inhibitor (IDN-6556) or by tumor-necrosis factor-α in combination with LCL-161 (Smac mimetic) and IDN-6556 (TSI). In these in vitro cellular models, celastrol inhibited the phosphorylation of RIPK1, RIPK3, and MLKL and the formation of necrosome during necroptotic induction, suggesting its possible action on upstream signaling of the necroptotic pathway. Consistent with the known role of mitochondrial dysfunction in necroptosis, we found that celastrol significantly rescued TSI-induced loss of mitochondrial membrane potential. TSI-induced intracellular and mitochondrial reactive oxygen species (mtROS), which are involved in the autophosphorylation of RIPK1 and recruitment of RIPK3, were significantly attenuated by celastrol. Moreover, in a mouse model of acute pancreatitis that is associated with necroptosis, celastrol administration significantly reduced the severity of caerulein-induced acute pancreatitis accompanied by decreased phosphorylation of MLKL in pancreatic tissues. Collectively, celastrol can attenuate the activation of RIPK1/RIPK3/MLKL signaling likely by attenuating mtROS production, thereby inhibiting necroptosis and conferring protection against caerulein-induced pancreatitis in mice.


Asunto(s)
Pancreatitis , Ratones , Animales , Pancreatitis/inducido químicamente , Pancreatitis/tratamiento farmacológico , Proteínas Quinasas/metabolismo , Necroptosis , Ceruletida , Enfermedad Aguda , Triterpenos Pentacíclicos , Caspasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Apoptosis
20.
Pharmacology ; 89(5-6): 348-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22699368

RESUMEN

AIM: The present study aimed to explore the antitumor effect and action mechanism of cucurbitacin B (CuB) on human T-cell leukemia Jurkat cells. METHODS: Cell proliferation was measured by the MTS assay. Cell cycle distribution, mitochondrial membrane potential and annexin V staining were analyzed using flow cytometry. Western blotting was used to determine the levels of apoptosis- and autophagy-related proteins. RESULTS: CuB inhibited the proliferation of Jurkat cells in a dose-dependent manner and induced G 2 /M phase arrest as well as formation of tetraploid cells. Accompanied with these effects, the actin dynamics was disrupted, and cofilin, a key regulator of actin dynamics, was persistently activated (dephosphorylated). Although CuB induced around 10% cells undergoing apoptosis, most of the cells were alive after CuB treatment for 24 h. Induction of autophagy was also evident by accumulation of LC3-II. CuB-induced autophagy seemed to be a prosurvival response, since suppression of CuB-induced autophagy significantly increased the activation of caspase-3. CONCLUSION: Our results demonstrated that CuB exhibited antitumor activity in Jurkat cells through induction of cell cycle arrest and apoptosis which was at least partly due to the disruption of actin dynamics.


Asunto(s)
Actinas/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Triterpenos/farmacología , Factores Despolimerizantes de la Actina/metabolismo , Autofagia/efectos de los fármacos , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Humanos , Células Jurkat , Potencial de la Membrana Mitocondrial/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA