Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.138
Filtrar
1.
Gen Physiol Biophys ; 43(3): 185-196, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38774919

RESUMEN

Ampelopsin (AMP) had a wound-healing effect in rat skin wounds with or without purulent infection. However, the role of AMP in diabetic wound healing remains poorly defined. Wounds were created on the dorsal skin of type 2 diabetic mouse model, and the histological features of wounds were examined by hematoxylin and eosin (HE) staining. Caspase-1 activity and the secretion of inflammatory cytokines were detected by enzyme-linked immunosorbent assay (ELISA). Cell viability and migration were examined through cell counting kit-8 (CCK-8) and wound healing assays, respectively. AMP facilitated wound healing in vivo. AMP notably facilitated platelet endothelial cell adhesion molecule-31 (CD31), collagen type I alpha 1 chain (COL1A1), and alpha-smooth muscle actin (α-SMA), and inhibited matrix metallopeptidase 9 (MMP9) and cyclooxygenase 2 (Cox2) expression in diabetic wounds. The inflammasome pathway was implicated in skin injury. AMP inhibited pro-inflammatory factor secretions and NLR family pyrin domain containing 3 (NLRP3) inflammasome pathway in diabetic wounds and high glucose-treated THP-1 macrophages. AMP-mediated NLRP3 inflammasome inhibition in THP-1 macrophages increased cell viability and migratory capacity in HaCaT cells. AMP facilitated diabetic wound healing and increased keratinocyte cell viability and migratory ability by inhibiting the NLRP3 inflammasome pathway in macrophages.


Asunto(s)
Inflamasomas , Queratinocitos , Macrófagos , Proteína con Dominio Pirina 3 de la Familia NLR , Cicatrización de Heridas , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/efectos de los fármacos , Ratones , Humanos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Masculino , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Transducción de Señal/efectos de los fármacos , Ratones Endogámicos C57BL , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células THP-1 , Células HaCaT , Flavonoides
2.
Mol Biol Rep ; 51(1): 655, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38739285

RESUMEN

BACKGROUND: There is limited data regarding the hazardous effect of gentamicin (GM) on the uterus and whether or not vinpocetine (Vinpo) ameliorates it. The present study aimed to identify the possible protective effect of Vinpo in GM-induced uterine injury in rats. METHODS: Female rats were assorted in control-group, Vinpo-group, GM-group, and Vinpo plus GM group. Serum and uterine GM concentration were measured. Uterine oxidative stress parameters besides inflammatory and apoptotic biomarkers were evaluated. Uterine histopathological examination and interlukin-1beta (IL-1ß) immune-histochemical study were detected. RESULTS: GM significantly increased uterine oxidative stress, inflammatory and apoptotic biomarkers. Histopathological picture of uterine damage and increased IL-1ß immunoexpression were detected. Vinpo significantly ameliorated the distributed GM concentration, oxidative stress, inflammatory and apoptotic biomarkers with a prompt improvement in histopathological picture and a decrease in IL-1ß immunoexpression. CONCLUSION: Vinpo protective effect against GM-induced uterine injury involves modulation of inflammasome/caspase-1/IL-1ß signaling pathway.


Asunto(s)
Caspasa 1 , Gentamicinas , Inflamasomas , Interleucina-1beta , Estrés Oxidativo , Transducción de Señal , Útero , Alcaloides de la Vinca , Animales , Femenino , Interleucina-1beta/metabolismo , Alcaloides de la Vinca/farmacología , Ratas , Caspasa 1/metabolismo , Gentamicinas/efectos adversos , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Útero/efectos de los fármacos , Útero/metabolismo , Útero/patología , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos
3.
Exp Cell Res ; 438(2): 114061, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38692345

RESUMEN

Acute myocardial infarction (AMI) is a prevalent cardiovascular disease with high morbidity and mortality rates worldwide. Pyroptosis is an inflammatory form of programmed cell death that has been linked to various pathological conditions. However, its exact contribution to the onset and progression of heart injury in AMI has not yet fully elucidated. Herein, we established mouse AMI model by ligating the left anterior descending artery and performed transcriptome analysis during the early phase of AMI. Mouse HL-1 and human AC-16 cardiomyocytes were subjected to hypoxia to simulate ischemic injury in vitro. Our results revealed a significant activation of the inflammatory response at 3 h post-ligation, as confirmed by RNA sequencing. We identified the occurrence of NLRP3 inflammasome-mediated pyroptosis in the cardiac tissues of human cases with AMI, as well as in mouse models of AMI and hypoxia-induced cardiomyocytes, using immunohistochemistry staining and Western blotting assays. Concurrently, pharmacological inhibition of NLRP3 inflammasome-mediated pyroptosis with MCC950 and VX-765 effectively decreased hypoxia-induced cardiomyocytes injury, while mitigating myocardial oxidative stress, apoptosis and inflammation caused by hypoxia. Moreover, the circulating levels of gasdermin D (GSDMD), the pyroptosis executor, were remarkably elevated in the plasma of mice with early AMI and in the supernatant of hypoxia-exposed cardiomyocytes in a time-dependent manner using ELISA and Western blotting. Furthermore, the change in circulating GSDMD positively correlated with Creatine Kinase-MB (CK-MB) in the plasma of early-stage AMI mouse. In summary, these findings indicated a critical role for NLRP3 inflammasome-mediated pyroptosis in the progression of AMI, the administration of MCC950 and VX-765 may be attractive candidate therapeutic approaches for cardiac injury caused by acute hypoxia or even AMI. Additionally, the circulating GSDMD exhibits potential as a newly diagnostic biomarker for AMI.


Asunto(s)
Apoptosis , Furanos , Inflamación , Ratones Endogámicos C57BL , Infarto del Miocardio , Miocitos Cardíacos , Estrés Oxidativo , Piroptosis , Sulfonamidas , Piroptosis/efectos de los fármacos , Animales , Ratones , Apoptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Sulfonamidas/farmacología , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/tratamiento farmacológico , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Masculino , Furanos/farmacología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/tratamiento farmacológico , Indenos/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , para-Aminobenzoatos/farmacología , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Modelos Animales de Enfermedad , Miocardio/metabolismo , Miocardio/patología , Hipoxia/metabolismo , Hipoxia/complicaciones , Dipéptidos
4.
Exp Biol Med (Maywood) ; 249: 10104, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38708425

RESUMEN

Seawater-drowning-induced acute lung injury (SD-ALI) is a life-threatening disorder characterized by increased alveolar-capillary permeability, an excessive inflammatory response, and refractory hypoxemia. Perfluorocarbons (PFCs) are biocompatible compounds that are chemically and biologically inert and lack toxicity as oxygen carriers, which could reduce lung injury in vitro and in vivo. The aim of our study was to explore whether the vaporization of PFCs could reduce the severity of SD-ALI in canines and investigate the underlying mechanisms. Eighteen beagle dogs were randomly divided into three groups: the seawater drowning (SW), perfluorocarbon (PFC), and control groups. The dogs in the SW group were intratracheally administered seawater to establish the animal model. The dogs in the PFC group were treated with vaporized PFCs. Probe-based confocal laser endomicroscopy (pCLE) was performed at 3 h. The blood gas, volume air index (VAI), pathological changes, and wet-to-dry (W/D) lung tissue ratios were assessed. The expression of heme oxygenase-1 (HO-1), nuclear respiratory factor-1 (NRF1), and NOD-like receptor family pyrin domain containing-3 (NLRP3) inflammasomes was determined by means of quantitative real-time polymerase chain reaction (qRT-PCR) and immunological histological chemistry. The SW group showed higher lung injury scores and W/D ratios, and lower VAI compared to the control group, and treatment with PFCs could reverse the change of lung injury score, W/D ratio and VAI. PFCs deactivated NLRP3 inflammasomes and reduced the release of caspase-1, interleukin-1ß (IL-1ß), and interleukin-18 (IL-18) by enhancing the expression of HO-1 and NRF1. Our results suggest that the vaporization of PFCs could attenuate SD-ALI by deactivating NLRP3 inflammasomes via the HO-1/NRF1 pathway.


Asunto(s)
Lesión Pulmonar Aguda , Fluorocarburos , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Fluorocarburos/farmacología , Perros , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Agua de Mar , Masculino , Ahogamiento/metabolismo , Modelos Animales de Enfermedad , Pulmón/patología , Pulmón/metabolismo , Pulmón/efectos de los fármacos
5.
Front Immunol ; 15: 1344098, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38711511

RESUMEN

Inflammatory responses, especially chronic inflammation, are closely associated with many systemic diseases. There are many ways to treat and alleviate inflammation, but how to solve this problem at the molecular level has always been a hot topic in research. The use of nanoparticles (NPs) as anti-inflammatory agents is a potential treatment method. We synthesized new hollow cerium oxide nanomaterials (hCeO2 NPs) doped with different concentrations of Cu5.4O NPs [the molar ratio of Cu/(Ce + Cu) was 50%, 67%, and 83%, respectively], characterized their surface morphology and physicochemical properties, and screened the safe concentration of hCeO2@Cu5.4O using the CCK8 method. Macrophages were cultured, and P.g-lipopolysaccharide-stimulated was used as a model of inflammation and co-cultured with hCeO2@Cu5.4O NPs. We then observe the effect of the transcription levels of CTSB, NLRP3, caspase-1, ASC, IL-18, and IL-1ß by PCR and detect its effect on the expression level of CTSB protein by Western blot. The levels of IL-18 and IL-1ß in the cell supernatant were measured by enzyme-linked immunosorbent assay. Our results indicated that hCeO2@Cu5.4O NPs could reduce the production of reactive oxygen species and inhibit CTSB and NLRP3 to alleviate the damage caused by the inflammatory response to cells. More importantly, hCeO2@Cu5.4O NPs showed stronger anti-inflammatory effects as Cu5.4O NP doping increased. Therefore, the development of the novel nanomaterial hCeO2@Cu5.4O NPs provides a possible new approach for the treatment of inflammatory diseases.


Asunto(s)
Antiinflamatorios , Cerio , Cobre , Inflamación , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Cerio/farmacología , Cerio/química , Transducción de Señal/efectos de los fármacos , Animales , Ratones , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Antiinflamatorios/farmacología , Nanopartículas , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo
6.
ACS Appl Mater Interfaces ; 16(19): 24295-24307, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38697643

RESUMEN

Pyroptosis has garnered increasing attention because of its ability to trigger robust antitumor immunity. Pyroptosis is initiated by the activation of inflammasomes, which are regulated by various organelles. The collaboration among organelles offers several protective mechanisms to prevent activation of the inflammasome, thereby limiting the induction of efficient pyroptosis. Herein, a multiorganelle homeostasis disruptor (denoted BLL) is constructed by encapsulating liposomes and bortezomib (BTZ) within a layered double hydroxide (LDH) nanocage to continuously activate inflammasomes for inducing efficient pyroptosis. In lysosomes, the negatively charged liposomes are released to recruit the NLRP3 inflammasomes through electrostatic interactions. ER stress is induced by BTZ to enhance the activation of the NLRP3 inflammasome. Meanwhile, the BLL nanocage exhibited H+-scavenging ability due to the weak alkalinity of LDH, thus disrupting the homeostasis of the lysosome and alleviating the degradation of the NLRP3 inflammasome by lysosomal-associated autophagy. Our results suggest that the BLL nanocage induces homeostatic imbalance in various organelles and efficient pyroptosis. We hope this work can provide new insights into the design of an efficient pyroptosis inducer by disrupting the homeostatic balance of multiple organelles and promote the development of novel antineoplastic platforms.


Asunto(s)
Homeostasis , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Piroptosis/efectos de los fármacos , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Homeostasis/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Humanos , Ratones , Bortezomib/farmacología , Bortezomib/química , Liposomas/química , Animales , Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Hidróxidos/química , Hidróxidos/farmacología , Nanoestructuras/química , Nanopartículas/química
7.
Life Sci ; 348: 122688, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38710284

RESUMEN

Coenzyme Q10 (CoQ10) occurs naturally in the body and possesses antioxidant and cardioprotective effects. Cardiotoxicity has emerged as a serious effect of the exposure to cadmium (Cd). This study investigated the curative potential of CoQ10 on Cd cardiotoxicity in mice, emphasizing the involvement of oxidative stress (OS) and NF-κB/NLRP3 inflammasome axis. Mice received a single intraperitoneal dose of CdCl2 (6.5 mg/kg) and a week after, CoQ10 (100 mg/kg) was supplemented daily for 14 days. Mice that received Cd exhibited cardiac injury manifested by the elevated circulating cardiac troponin T (cTnT), CK-MB, LDH and AST. The histopathological and ultrastructural investigations supported the biochemical findings of cardiotoxicity in Cd-exposed mice. Cd administration increased cardiac MDA, NO and 8-oxodG while suppressed GSH and antioxidant enzymes. CoQ10 decreased serum CK-MB, LDH, AST and cTnT, ameliorated histopathological and ultrastructural changes in the heart of mice, decreased cardiac MDA, NO, and 8-OHdG and improved antioxidants. CoQ10 downregulated NF-κB p65, NLRP3 inflammasome, IL-1ß, MCP-1, JNK1, and TGF-ß in the heart of Cd-administered mice. Moreover, in silico molecular docking revealed the binding potential between CoQ10 and NF-κB, ASC1 PYD domain, NLRP3 PYD domain, MCP-1, and JNK. In conclusion, CoQ10 ameliorated Cd cardiotoxicity by preventing OS and inflammation and modulating NF-κB/NLRP3 inflammasome axis in mice. Therefore, CoQ10 exhibits potent therapeutic benefits in safeguarding cardiac tissue from the harmful consequences of exposure to Cd.


Asunto(s)
Cadmio , Cardiotoxicidad , Inflamasomas , FN-kappa B , Proteína con Dominio Pirina 3 de la Familia NLR , Estrés Oxidativo , Ubiquinona , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratones , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Estrés Oxidativo/efectos de los fármacos , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/metabolismo , Cardiotoxicidad/prevención & control , FN-kappa B/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Masculino , Cadmio/toxicidad , Regulación hacia Abajo/efectos de los fármacos , Antioxidantes/farmacología
8.
Toxicol Appl Pharmacol ; 486: 116952, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38705399

RESUMEN

The incidence of contrast-induced acute kidney injury (CI-AKI) has escalated to become the third most prevalent cause of hospital-acquired AKI, with a lack of efficacious interventions. Berberine (BBR) possesses diverse pharmacological effects and exhibits renoprotective properties; however, limited knowledge exists regarding its impact on CI-AKI. Therefore, our study aimed to investigate the protective effects and underlying mechanisms of BBR on CI-AKI in a mice model, focusing on the nucleotide-binding oligomerization domain-like pyrin domain-containing protein 3 (NLRP3) inflammasome and mitophagy. The CI-AKI mice model was established by administering NG-nitro-L-arginine methyl ester (L-NAME) (10 mg/kg), indomethacin (10 mg/kg), and iohexol (11 g/kg) following water deprivation. A pretreatment of 100 mg/kg of BBR was orally administered to the mice for two weeks. Renal injury markers, damage-associated molecular patterns (DAMPs), renal histopathology, mitochondrial morphology, autophagosomes, and potential mechanisms were investigated. BBR effectively reduced levels of renal injury biomarkers such as serum cystatin C, urea nitrogen, and creatinine, downregulated the protein level of kidney injury molecule 1 (KIM1), and mitigated renal histomorphological damage. Moreover, BBR reduced DAMPs, including high mobility group box-1 (HMGB1), heat shock protein 70 (HSP70), and uric acid (UA). It also alleviated oxidative stress and inflammatory factors such as monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1ß). Furthermore, the activation of NLRP3 inflammasome was attenuated in the BBR pretreatment group, as evidenced by both mRNA and protein levels. Electron microscopy and western blotting examination revealed that BBR mitigated mitochondrial damage and enhanced mitophagy. Additionally, BBR increased the P-AMPK/AMPK ratio. These findings indicated that BBR exerted a protective effect against CI-AKI by suppressing NLRP3 inflammasome activation and modulating mitophagy, providing a potential therapeutic strategy for its prevention.


Asunto(s)
Lesión Renal Aguda , Berberina , Medios de Contraste , Modelos Animales de Enfermedad , Inflamasomas , Ratones Endogámicos C57BL , Mitofagia , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Mitofagia/efectos de los fármacos , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/tratamiento farmacológico , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Ratones , Berberina/farmacología , Masculino , Riñón/efectos de los fármacos , Riñón/patología , Riñón/metabolismo
10.
Ecotoxicol Environ Saf ; 276: 116287, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38579532

RESUMEN

Benzo(a)pyrene (BaP) can be detected in the human placenta. However, little is known about the effects of BaP exposure on different placental cells under various conditions. In this study, we aimed to investigate the effects of BaP on mitochondrial function, pyrin domain-containing protein 3 (NLRP3) inflammasome, and apoptosis in three human trophoblast cell lines under normoxia, hypoxia, and inflammatory conditions. JEG-3, BeWo, and HTR-8/SVneo cell lines were exposed to BaP under normoxia, hypoxia, or inflammatory conditions for 24 h. After treatment, we evaluated cell viability, apoptosis, aryl hydrocarbon receptor (AhR) protein and cytochrome P450 (CYP) gene expression, mitochondrial function, including mitochondrial DNA copy number (mtDNAcn), mitochondrial membrane potential (ΔΨm), intracellular adenosine triphosphate (iATP), and extracellular ATP (eATP), nitric oxide (NO), NLPR3 inflammasome proteins, and interleukin (IL)-1ß. We found that BaP upregulated the expression of AhR or CYP genes to varying degrees in all three cell lines. Exposure to BaP alone increased ΔΨm in all cell lines but decreased NO in BeWo and HTR-8/SVneo, iATP in HTR-8/SVneo, and cell viability in JEG-3, without affecting apoptosis. Under hypoxic conditions, BaP did not increase the expression of AhR and CYP genes in JEG-3 cells but increased CYP gene expression in two others. Pro-inflammatory conditions did not affect the response of the 3 cell lines to BaP with respect to the expression of CYP genes and changes in the mitochondrial function and NLRP3 inflammasome proteins. In addition, in HTR-8/SVneo cells, BaP increased IL-1ß secretion in the presence of hypoxia and poly(I:C). In conclusion, our results showed that BaP affected mitochondrial function in trophoblast cell lines by increasing ΔΨm. This increased ΔΨm may have rescued the trophoblast cells from activation of the NLRP3 inflammasome and apoptosis after BaP treatment. We also observed that different human trophoblast cell lines had cell type-dependent responses to BaP exposure under normoxia, hypoxia, or pro-inflammatory conditions.


Asunto(s)
Apoptosis , Benzo(a)pireno , Supervivencia Celular , Proteína con Dominio Pirina 3 de la Familia NLR , Placenta , Receptores de Hidrocarburo de Aril , Trofoblastos , Humanos , Benzo(a)pireno/toxicidad , Placenta/efectos de los fármacos , Placenta/citología , Línea Celular , Femenino , Embarazo , Apoptosis/efectos de los fármacos , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Supervivencia Celular/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Mitocondrias/efectos de los fármacos , Inflamación/inducido químicamente , Hipoxia de la Célula/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética
11.
Mar Drugs ; 22(4)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38667787

RESUMEN

Porphyran, a sulfated polysaccharide found in various species of marine red algae, has been demonstrated to exhibit diverse bioactivities, including anti-inflammatory effects. However, the protective effects of porphyran against cerebral ischemia and reperfusion (IR) injury have not been investigated. The aim of this study was to examine the neuroprotective effects of porphyran against brain IR injury and its underlying mechanisms using a gerbil model of transient forebrain ischemia (IR in the forebrain), which results in pyramidal cell (principal neuron) loss in the cornu ammonis 1 (CA1) subregion of the hippocampus on day 4 after IR. Porphyran (25 and 50 mg/kg) was orally administered daily for one week prior to IR. Pretreatment with 50 mg/kg of porphyran, but not 25 mg/kg, significantly attenuated locomotor hyperactivity and protected pyramidal cells located in the CA1 area from IR injury. The pretreatment with 50 mg/kg of porphyran significantly suppressed the IR-induced activation and proliferation of microglia in the CA1 subregion. Additionally, the pretreatment significantly inhibited the overexpressions of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing protein-3 (NLRP3) inflammasome complex, and pro-inflammatory cytokines (interleukin 1 beta and interleukin 18) induced by IR in the CA1 subregion. Overall, our findings suggest that porphyran exerts neuroprotective effects against brain IR injury, potentially by reducing the reaction (activation) and proliferation of microglia and reducing NLRP3 inflammasome-mediated neuroinflammation.


Asunto(s)
Región CA1 Hipocampal , Gerbillinae , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Enfermedades Neuroinflamatorias , Fármacos Neuroprotectores , Daño por Reperfusión , Sefarosa/análogos & derivados , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Masculino , Daño por Reperfusión/tratamiento farmacológico , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Modelos Animales de Enfermedad , Microglía/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Polisacáridos/farmacología , Neuronas/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo
12.
J Cell Mol Med ; 28(9): e18338, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38683122

RESUMEN

Respiratory syncytial virus (RSV) infects neuronal cells in the central nervous system (CNS), resulting in neurological symptoms. In the present study, we intended to explore the mechanism of RSV infection-induced neuroinflammatory injury from the perspective of the immune response and sought to identify effective protective measures against the injury. The findings showed that toll-like receptor 4 (TLR4) was activated after RSV infection in human neuronal SY5Y cells. Furthermore, TLR4 activation induced autophagy and apoptosis in neuronal cells, promoted the formation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, and increased the secretion of downstream inflammatory cytokines such as interleukin-1ß (IL-1ß), interleukin-18 (IL-18) and tumour necrosis factor-α (TNF-α). Interestingly, blockade of TLR4 or treatment with exogenous melatonin significantly suppressed TLR4 activation as well as TLR4-mediated apoptosis, autophagy and immune responses. Therefore, we infer that melatonin may act on the TLR4 to ameliorate RSV-induced neuronal injury, which provides a new therapeutic target for RSV infection.


Asunto(s)
Apoptosis , Autofagia , Inflamasomas , Melatonina , Proteína con Dominio Pirina 3 de la Familia NLR , Infecciones por Virus Sincitial Respiratorio , Receptor Toll-Like 4 , Humanos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Sistema Nervioso Central/virología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/patología , Citocinas/metabolismo , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Melatonina/farmacología , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/virología , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/fisiología , Receptor Toll-Like 4/efectos de los fármacos , Receptor Toll-Like 4/metabolismo
13.
Exp Cell Res ; 438(1): 114052, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38636651

RESUMEN

Trained immunity is mechanistically defined as the metabolically and epigenetically mediated long-term functional adaptation of the innate immune system, characterized by a heightened response to a secondary stimulation. Given appropriate activation, trained immunity represents an attractive anti-infective therapeutic target. Nevertheless, excessive immune response and subsequent inflammatory cascades may contribute to pathological tissue damage, indicating that the negative impacts of trained immunity appear to be significant. In this study, we show that innate immune responses such as the production of extracellular traps, pro-inflammatory cytokines, and autophagy-related proteins were markedly augmented in trained BMDMs. Furthermore, heat-killed C. albicans priming promotes the activation of the AIM2 inflammasome, and AIM2-/- mice exhibit impaired memory response induced by heat-killed C. albicans. Therefore, we establish that the AIM2 inflammasome is involved in trained immunity and emerges as a promising therapeutic target for potentially deleterious effects. Dihydroartemisinin can inhibit the memory response induced by heat-killed C. albicans through modulation of mTOR signaling and the AIM2 inflammasome. The findings suggest that dihydroartemisinin can reduce the induction of trained immunity by heat-killed C. albicans in C57BL/6 mice. Dihydroartemisinin is one such therapeutic intervention that has the potential to treat of diseases characterized by excessive trained immunity.


Asunto(s)
Artemisininas , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal/efectos de los fármacos , Ratones , Artemisininas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Candida albicans/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Ratones Noqueados , Inmunidad Entrenada
14.
Mol Biol Rep ; 51(1): 593, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38683404

RESUMEN

BACKGROUND: Parkinson's disease (PD) is a common central nervous system neurodegenerative disease. Neuroinflammation is one of the significant neuropathological hallmarks. As a traditional Chinese medicine, Safranal exerts anti-inflammatory effects in various diseases, however, whether it plays a similar effect on PD is still unclear. The study was to investigate the effects and mechanism of Safranal on PD. METHODS: The PD mouse model was established by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine MPTP firstly. Next, the degree of muscle stiffness, neuromuscular function, motor retardation and motor coordination ability were examined by observing and testing mouse movement behavior. Immunofluorescence staining was used to observe the expression of tyrosine hydroxylase (TH). The dopamine (DA) content of the striatum was detected by High-performance liquid chromatography (HPLC). The expression of TH and NLRP3 inflammasome-related markers NLRP3, IL-1ß, and Capase-1 were detected by Real-time Polymerase Chain Reaction (qRT-PCR) and western blotting (WB) respectively. RESULTS: Through behavioral testing, Parkinson's mouse showed a higher muscle stiffness and neuromuscular tension, a more motor retardation and activity disorders, together with a worse motor coordination compared with sham group. Simultaneously, DA content and TH expression in the striatum were decreased. However, after using Safranal treatment, the above pathological symptoms of Parkinson's mouse all improved compared with Safranal untreated group, the DA content and TH expression were also increased to varying degrees. Surprisingly, it observed a suppression of NLRP3 inflammation in the striatum of Parkinson's mouse. CONCLUSIONS: Safranal played a neuroprotective effect on the Parkinson's disease and its mechanism was related to the inhibition of NLRP3 inflammasome activation.


Asunto(s)
Ciclohexenos , Modelos Animales de Enfermedad , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Fármacos Neuroprotectores , Enfermedad de Parkinson , Terpenos , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratones , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Terpenos/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Masculino , Ciclohexenos/farmacología , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Ratones Endogámicos C57BL , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Dopamina/metabolismo , Cuerpo Estriado/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Interleucina-1beta/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Caspasa 1/metabolismo
15.
Neuropharmacology ; 252: 109950, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38636727

RESUMEN

Effective therapeutic interventions for elderly patients are lacking, despite advances in pharmacotherapy. Methylated urolithin A (mUro A), a modified ellagitannin (ET)-derived metabolite, exhibits anti-inflammatory, antioxidative, and anti-apoptotic effects. Current research has primarily investigated the neuroprotective effects of mUroA in aging mice and explored the underlying mechanisms. Our study used an in vivo aging model induced by d-galactose (D-gal) to show that mUro A notably improved learning and memory, prevented synaptic impairments by enhancing synaptic protein expression and increasing EPSCs, and reduced oxidative damage in aging mice. mUro A alleviated the activation of the NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome, leading to reduced glial cell activity and neuroinflammation in both accelerated aging and naturally senescent mouse models. Moreover, mUroA enhanced the activity of TCA cycle enzymes (PDH, CS, and OGDH), decreased 8-OHdG levels, and raised ATP and NAD+ levels within the mitochondria. At the molecular level, mUro A decreased phosphorylated p53 levels and increased the expression of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), thus enhancing mitochondrial function. In conclusion, mUro A alleviates cognitive impairment in aging mice by suppressing neuroinflammation through NLRP3 inflammasome inhibition and restoring mitochondrial function via the p53-PGC-1α pathway. This suggests its potential therapeutic agent for brain aging and aging-related diseases.


Asunto(s)
Envejecimiento , Disfunción Cognitiva , Cumarinas , Inflamasomas , Ratones Endogámicos C57BL , Mitocondrias , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Cumarinas/farmacología , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ratones , Masculino , Galactosa , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo
16.
Gene ; 919: 148509, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38677349

RESUMEN

BACKGROUD: It is widely acknowledged that Metformin (MET), an established medication for managing type 2 diabetes, possesses diverse pharmacological effects. This study aims to investigate the protective effects of MET against Nω-Nitro-L-arginine methyl ester (L-NAME)-induced preeclampsia (PE). METHODS: Sprague Dawley (SD) rats were exposed to 200 mg/kg L-NAME with or without prior MET treatment. Histopathological analysis was performed using Hematoxylin and Eosin staining. Serum levels of inflammatory, antiangiogenic, and angiogenic factors were quantified using ELISA kits. Immunohistochemistry (IHC) staining was employed to observe NLRP3 and IL-1ß expressions in placental tissues. Western blot and Quantitative Real-Time PCR (q-PCR) analyses were conducted to assess protein and mRNA expressions of NLRP3, caspase-1, ASC, and IL-1ß. RESULTS: We found that MET could mitigate placental histopathological deterioration and improve pregnancy outcomes in L-NAME-induced PE rat models. MET not only suppressed L-NAME-induced elevation of antiangiogenic factors but also stimulated the production of pro-angiogenic factors. Additionally, MET treatment reversed the excessive inflammatory response induced by L-NAME. Furthermore, MET inhibited the activation of the NLRP3 inflammasome triggered by L-NAME, evidenced by the downregulation of NLRP3 expression, caspase-1, and IL-1ß. CONCLUSIONS: MET demonstrates a protective effect against L-NAME-induced PE rats, potentially mediated through inhibition of the inflammatory response, downregulation of NLRP3 inflammasome expression in the placenta, and regulation of the balance between anti-angiogenic and pro-angiogenic factors.


Asunto(s)
Inflamasomas , Metformina , NG-Nitroarginina Metil Éster , Proteína con Dominio Pirina 3 de la Familia NLR , Placenta , Preeclampsia , Ratas Sprague-Dawley , Animales , Preeclampsia/tratamiento farmacológico , Preeclampsia/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Femenino , Metformina/farmacología , Embarazo , Ratas , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Placenta/metabolismo , Placenta/efectos de los fármacos , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Modelos Animales de Enfermedad , Caspasa 1/metabolismo
17.
Biomed Pharmacother ; 174: 116548, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38599064

RESUMEN

BACKGROUND: Various heart diseases ultimately lead to chronic heart failure (CHF). In CHF, the inflammatory response is associated with pyroptosis, which is mediated by the NOD-like receptor protein 3 (NLRP3) inflammasome. Fu Xin decoction (FXD) is commonly used in clinical practice to treat CHF and improve inflammatory conditions. However, the specific pharmacological mechanisms of action for FXD in these processes have yet to be fully understood. PURPOSE: The objective of this study was to examine the protective mechanism of FXT against CHF, both in H9c2 cells and mice. METHOD: A CHF mouse model was established, and the effect of FXD was observed via gavage. Cardiac function was evaluated using echocardiography, while serum BNP and LDH levels were analyzed to assess the severity of CHF. Hematoxylin and eosin staining (H&E) and Masson staining were performed to evaluate myocardial pathological changes, and TdT-mediated dUTP Nick-End Labeling staining was used to detect DNA damage. Additionally, doxorubicin was utilized to induce myocardial cell injury in H9c2 cells, establishing a relevant model. CCK8 was used to observe cell viability and detect LDH levels in the cell supernatant. Subsequently, the expression of pyroptosis-related proteins was detected using immunohistochemistry, immunofluorescence, and western blotting. Finally, the pharmacological mechanism of FXD against CHF was further validated by treating H9c2 cells with an NLRP3 activator and inducing NLRP3 overexpression. RESULT: According to current research findings, echocardiography demonstrated a significant improvement of cardiac function by FXD, accompanied by reduced levels of BNP and LDH, indicating the amelioration of cardiac injury in CHF mice. FXD exhibited the ability to diminish serum CRP and MCP inflammatory markers in CHF mice. The results of HE and Masson staining analyses revealed a significant reduction in pathological damage of the heart tissue following FXD treatment. The CCK8 assay demonstrated the ability of FXD to enhance H9c2 cell viability, improve cell morphology, decrease LDH levels in the cell supernatant, and alleviate cell damage. Immunohistochemistry, Western blotting, and immunofluorescence staining substantiated the inhibitory effect of FXD on the NLRP3/caspase-1/GSDMD pyroptosis signaling pathway in both CHF and H9c2 cell injury models. Ultimately, the administration of the NLRP3 activator (Nigericin) and the overexpression of NLRP3 counteract the effects of FXD on cardiac protection and pyroptosis inhibition in vitro. CONCLUSION: FXD exhibits a cardioprotective effect, improving CHF and alleviating pyroptosis by inhibiting the NLRP3/caspase-1/GSDMD pathway.


Asunto(s)
Medicamentos Herbarios Chinos , Insuficiencia Cardíaca , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Animales , Ratones , Caspasa 1/efectos de los fármacos , Caspasa 1/metabolismo , Línea Celular , Enfermedad Crónica , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Gasderminas/efectos de los fármacos , Gasderminas/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Piroptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
18.
Int Immunopharmacol ; 133: 112099, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38643709

RESUMEN

Visceral hypersensitivity resulting from compromised gut barrier with activated immune system is a key feature of irritable bowel syndrome (IBS). Corticotropin-releasing factor (CRF) and Toll-like receptor 4 (TLR4) activate proinflammatory cytokine signaling to induce these changes, which is one of the mechanisms of IBS. As activation of the NLRP3 inflammasome by lipopolysaccharide (LPS) or TLR4 leads to release interleukin (IL)-1ß, the NLRP3 inflammasome may be involved in the pathophysiology of IBS. Tranilast, an anti-allergic drug has been demonstrated to inhibit the NLRP3 inflammasome, and we evaluated the impact of tranilast on visceral hypersensitivity and colonic hyperpermeability induced by LPS or CRF (IBS rat model). Visceral pain threshold caused by colonic balloon distention was measured by monitoring abdominal muscle contractions electrophysiologically. Colonic permeability was determined by quantifying the absorbed Evans blue within the colonic tissue. Colonic protein levels of NLRP3 and IL-1ß were assessed by immunoblot or ELISA. Intragastric administration of tranilast (20-200 mg/kg) for 3 days inhibited LPS (1 mg/kg)-induced visceral hypersensitivity and colonic hyperpermeability in a dose-dependent manner. Simultaneously, tranilast also abolished these alterations induced by CRF (50 µg/kg). LPS increased colonic protein levels of NLRP3 and IL-1ß, and tranilast inhibited these changes. ß-hydroxy butyrate, an NLRP3 inhibitor, also abolished visceral hypersensitivity and colonic hyperpermeability caused by LPS. In contrast, IL-1ß induced similar GI alterations to LPS, which were not modified by tranilast. In conclusion, tranilast improved visceral pain and colonic barrier by suppression of the NLRP3 inflammasome in IBS rat models. Tranilast may be useful for IBS treating.


Asunto(s)
Colon , Modelos Animales de Enfermedad , Inflamasomas , Interleucina-1beta , Síndrome del Colon Irritable , Lipopolisacáridos , Proteína con Dominio Pirina 3 de la Familia NLR , Ratas Sprague-Dawley , ortoaminobenzoatos , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Síndrome del Colon Irritable/tratamiento farmacológico , Síndrome del Colon Irritable/metabolismo , Colon/efectos de los fármacos , Colon/metabolismo , Masculino , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , ortoaminobenzoatos/farmacología , ortoaminobenzoatos/uso terapéutico , Interleucina-1beta/metabolismo , Ratas , Permeabilidad/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Dolor Visceral/tratamiento farmacológico , Dolor Visceral/metabolismo
19.
Mol Nutr Food Res ; 68(9): e2400048, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38659317

RESUMEN

Egg yolk lipids significantly alleviate dextran sulfate sodium (DSS)-induced colitis by inhibiting NLRP3 inflammasome, reversing gut microbiota dysbiosis, and increasing short chain fatty acids (SCFAs) concentrations. However, the role of gut microbiota and the relationship between SCFAs and NLRP3 inflammasome are still unknown. Here, this study confirms that antibiotic treatment abolishes the protective effect of egg yolk lipids on DSS-induced colonic inflammation, intestinal barrier damage, and lipopolysaccharide translocation. Fecal microbiota transplantation also supports that egg yolk lipids alleviate colitis in a gut microbiota-dependent manner. Then, the study investigates the relationship between SCFAs and NLRP3 inflammasome, and finds that SCFAs significantly suppress colitis via inhibiting colonic NLRP3 inflammasome activation and proinflammatory cytokines secretions (interleukin, IL)-1ß and IL-18, and combined treatment of SCFAs and MCC950 (NLRP3 inhibitor) shows a better activity against colitis and NLRP3 inflammasome activation. Together, these findings provide positive evidence for gut microbiorta-SCFAs-NLRP3 axis as a novel target involving in the therapy of inflammatory bowel disease.


Asunto(s)
Colitis , Sulfato de Dextran , Yema de Huevo , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Inflamasomas , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ácidos Grasos Volátiles/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Masculino , Ratones , Trasplante de Microbiota Fecal , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Lípidos , Interleucina-1beta/metabolismo
20.
J Affect Disord ; 356: 586-596, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38657764

RESUMEN

BACKGROUND: Diabetes mellitus (DM) is frequently associated with the occurrence and development of depression, and the co-occurrence of diabetes mellitus with depression (DD) may further reduce patients' quality of life. Recent research indicates that dopamine receptors (DRs) play a crucial role in immune and metabolic regulation. Pramipexole (PPX), a D2/3R agonist, has demonstrated promising neuroprotective and immunomodulatory effects. Nevertheless, the therapeutic effects and mechanisms of action of PPX on DM-induced depression are not clear at present. METHODS: Depression, DM, and DD were induced in a rat model through a combination of a high-fat diet (HFD) supplemented with streptozotocin (STZ) and chronic unpredictable mild stress (CUMS) combined with solitary cage rearing. The pathogenesis of DD and the neuroprotective effects of DRs agonists were investigated using behavioral assays, enzyme-linked immunosorbent assay (ELISA), hematoxylin-eosin (HE) staining, Nissl staining, Western blotting (WB) and immunofluorescence (IF). RESULTS: DD rats exhibited more severe dopaminergic, neuroinflammatory, and neuroplastic impairments and more pronounced depressive behaviors than rats with depression alone or DM. Our findings suggest that DRs agonists have significant therapeutic effects on DD rats and that PPX improved neuroplasticity and decreased neuroinflammation in the hippocampus of DD rats while also promoting DG cell growth and differentiation, ultimately mitigating depression-like behaviors. LIMITATION: Our study is based on a rat model. Further evidence is needed to determine whether the therapeutic effects of PPX apply to patients suffering from DD. CONCLUSIONS: Neuroinflammation mediated by damage to the dopaminergic system is one of the key pathogenic mechanisms of DD. We provide evidence that PPX has a neuroprotective effect on the hippocampus in DD rats and the mechanism may involve the inhibition of NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome activation by DRs to attenuate the neuroinflammatory response and neuroplasticity damage.


Asunto(s)
Depresión , Diabetes Mellitus Experimental , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Plasticidad Neuronal , Pramipexol , Animales , Pramipexol/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratas , Plasticidad Neuronal/efectos de los fármacos , Masculino , Inflamasomas/efectos de los fármacos , Depresión/tratamiento farmacológico , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/complicaciones , Ratas Sprague-Dawley , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Agonistas de Dopamina/farmacología , Hipocampo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...