Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 740
Filtrar
1.
J Cell Mol Med ; 28(12): e18458, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39031798

RESUMEN

Schistosomiasis is a parasitic disease characterized by liver fibrosis, a process driven by the activation of hepatic stellate cells (HSCs) and subsequent collagen production. Previous studies from our laboratory have demonstrated the ability of Schistosoma japonicum protein P40 (SjP40) to inhibit HSCs activation and exert an antifibrotic effect. In this study, we aimed to elucidate the molecular mechanism underlying the inhibitory effect of recombinant SjP40 (rSjP40) on HSCs activation. Using a cell model in which rSjP40 inhibited LX-2 cell activation, we performed RNA-seq analyses and identified ATF3 as the most significantly altered gene. Further investigation revealed that rSjP40 inhibited HSCs activation partly by suppressing ATF3 activation. Knockdown of ATF3 in mouse liver significantly alleviated S. japonicum-induced liver fibrosis. Moreover, our results indicate that ATF3 is a direct target of microRNA-494-3p, a microRNA associated with anti-liver fibrosis effects. rSjP40 was found to downregulate ATF3 expression by upregulating microRNA-494-3p in LX-2 cells. This downregulation led to the inhibition of the expression of liver fibrosis proteins α-SMA and COL1A1, ultimately alleviating liver fibrosis caused by S. japonicum.


Asunto(s)
Factor de Transcripción Activador 3 , Proteínas del Helminto , Células Estrelladas Hepáticas , Cirrosis Hepática , MicroARNs , Schistosoma japonicum , Esquistosomiasis Japónica , Animales , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/parasitología , Esquistosomiasis Japónica/parasitología , Esquistosomiasis Japónica/metabolismo , Esquistosomiasis Japónica/genética , Cirrosis Hepática/parasitología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Cirrosis Hepática/metabolismo , Ratones , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Actinas/metabolismo , Actinas/genética , Línea Celular , Regulación de la Expresión Génica , Hígado/metabolismo , Hígado/parasitología , Hígado/patología , Modelos Animales de Enfermedad , Antígenos Helmínticos
2.
Sci Adv ; 10(30): eado3141, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39047111

RESUMEN

Metabolic dysfunction-associated steatohepatitis (MASH) is regulated by complex interplay between the macrophages and surrounding cells in the liver. Here, we show that Atf3 regulates glucose-fatty acid cycle in macrophages attenuates hepatocyte steatosis, and fibrogenesis in hepatic stellate cells (HSCs). Overexpression of Atf3 in macrophages protects against the development of MASH in Western diet-fed mice, whereas Atf3 ablation has the opposite effect. Mechanistically, Atf3 improves the reduction of fatty acid oxidation induced by glucose via forkhead box O1 (FoxO1) and Cd36. Atf3 inhibits FoxO1 activity via blocking Hdac1-mediated FoxO1 deacetylation at K242, K245, and K262 and increases Zdhhc4/5-mediated CD36 palmitoylation at C3, C7, C464, and C466; furthermore, macrophage Atf3 decreases hepatocytes lipogenesis and HSCs activation via retinol binding protein 4 (Rbp4). Anti-Rbp4 can prevent MASH progression that is induced by Atf3 deficiency in macrophages. This study identifies Atf3 as a regulator of glucose-fatty acid cycle. Targeting macrophage Atf3 or Rbp4 may be a plausible therapeutic strategy for MASH.


Asunto(s)
Factor de Transcripción Activador 3 , Macrófagos , Animales , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Ratones , Macrófagos/metabolismo , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/etiología , Células Estrelladas Hepáticas/metabolismo , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Hígado/metabolismo , Hígado/patología , Hepatocitos/metabolismo , Antígenos CD36/metabolismo , Antígenos CD36/genética , Lipogénesis , Humanos , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Reprogramación Celular , Ratones Endogámicos C57BL , Reprogramación Metabólica
3.
Tissue Cell ; 89: 102447, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38991270

RESUMEN

Activating transcription factor 3 (ATF3) has been identified as a regulator associated with osteoblast differentiation. However, the effects of ATF3 on the osteogenic differentiation and proliferation of human periodontal stem cells (hPDLSCs) in periodontitis have not been reported. With the purpose of establishing an in vitro model of periodontitis, hPDLSCs were challenged with lipopolysaccharide (LPS). The Cell Counting Kit-8 assay was applied to assess cell viability, while reverse transcription-quantitative PCR and western blotting were employed to detect ATF3 expression. Inflammatory release was assessed using ELISA, together with western blotting. Lipid peroxidation was explored using the C11 BODIPY 581/591 probe, biochemical kits, thiobarbituric acid reactive substances (TBARS) assay and DCFH-DA staining. Iron and Fe2+ levels, and the expression levels of ferroptosis-related proteins were measured using corresponding kits and western blotting. Osteogenic differentiative capability was evaluated using alkaline phosphatase staining, Alizarin red staining and western blotting. The expression levels of proteins associated with Nrf2/HO-1 signaling were identified using western blotting. The results indicated that ATF3 expression was upregulated in LPS-induced hPDLSCs. The knockdown of ATF3 alleviated the LPS-induced inflammatory response in hPDLSCs, together with increased levels of TNF-α, IL-6, IL-1ß, Cox-2 and iNOS, and decreased levels of IL-10. ATF3 silencing also led to lower TBARS production rate, and reduced levels of reactive oxygen species, iron, Fe2+, ACSL4 and TFR1, whereas it elevated the levels of SLC7A11 and GPX4. In addition, ATF3 silencing promoted hPDLSC mineralization and cell differentiation, and elevated the levels of OCN2, RUNX2 and BMP2. Additionally, ATF3 depletion upregulated the expression levels of proteins related with Nrf2/HO-1 signaling. The Nrf2 inhibitor ML385 partially counteracted the effects of ATF3 interference on the LPS-challenged inflammatory response, lipid peroxidation, ferroptosis as well as osteogenic differentiative capability in hPDLSCs. In summary, the results revealed that ATF3 silencing suppressed inflammation and ferroptosis, while it improved osteogenic differentiation in LPS-induced hPDLSCs by regulating Nrf2/HO-1 signaling, which may provide promising therapeutic targets for the treatment of periodontitis.


Asunto(s)
Factor de Transcripción Activador 3 , Diferenciación Celular , Ferroptosis , Hemo-Oxigenasa 1 , Inflamación , Factor 2 Relacionado con NF-E2 , Osteogénesis , Transducción de Señal , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Inflamación/metabolismo , Inflamación/patología , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Lipopolisacáridos/farmacología , Células Madre/metabolismo
4.
Sci Rep ; 14(1): 14669, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918575

RESUMEN

Non-obese diabetes (NOD) mice are an established, spontaneous model of type 1 diabetes in which diabetes develops through insulitis. Using next-generation sequencing, coupled with pathway analysis, the molecular fingerprint of early insulitis was mapped in a cohort of mice ranging from 4 to 12 weeks of age. The resulting dynamic timeline revealed an initial decrease in proliferative capacity followed by the emergence of an inflammatory signature between 6 and 8 weeks that increased to a regulatory plateau between 10 and 12 weeks. The inflammatory signature is identified by the activation of central immunogenic factors such as Infg, Il1b, and Tnfa, and activation of canonical inflammatory signaling. Analysis of the regulatory landscape revealed the transcription factor Atf3 as a potential novel modulator of inflammatory signaling in the NOD islets. Furthermore, the Hedgehog signaling pathway correlated with Atf3 regulation, suggesting that the two play a role in regulating islet inflammation; however, further studies are needed to establish the nature of this connection.


Asunto(s)
Factor de Transcripción Activador 3 , Diabetes Mellitus Tipo 1 , Islotes Pancreáticos , Ratones Endogámicos NOD , Transducción de Señal , Animales , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Ratones , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Femenino , Inflamación/genética , Inflamación/patología , Inflamación/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Perfilación de la Expresión Génica , Modelos Animales de Enfermedad
5.
Gene ; 927: 148670, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38857714

RESUMEN

LINC00894 may be associated with synaptic function, but its biology function in neural cells is still unknown. In this study, LINC00894 knockdown decreased the EdU incorporated into newly synthesized DNA and cell viability in MTT or CCK-8 assay in HEK-293T and BE(2)-M17 (M17) neuroblastoma cells. And LINC00894 knockdown increased cellular apoptosis in Annexin V-FITC staining, the expression of activated Caspase3 and the level of reactive oxygen species (ROS) both in HEK-293T and M17 cells. Moreover, LINC00894 also protected cells from hydrogen peroxide induced apoptosis in in vitro models. Utilizing RNA sequencing (RNA-seq) integrated with quantitative reverse transcription polymerase chain reaction (RT-qPCR) and immunoblot, we identified that LINC00894 affected activating transcription factor 3 (ATF3) expression in HEK-293T, M17, and SH-SY5Y neuroblastoma cells. Finally, we found that ectopic expression of ATF3 restored cell proliferation and inhibited cell apoptosis in LINC00894 downregulated M17 cells. While knockdown of ATF3 also significantly increased the cell viability inhibition and apoptosis promotion induced by LINC00894 knockdown in M17 cells. Our results from in vitro models revealed that LINC00894 could promote neuronal cell proliferation and inhibit cellular apoptosis by affecting ATF3 expression.


Asunto(s)
Factor de Transcripción Activador 3 , Apoptosis , Proliferación Celular , Neuronas , ARN Largo no Codificante , Humanos , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Células HEK293 , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neuronas/metabolismo , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Supervivencia Celular , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Técnicas de Silenciamiento del Gen , Caspasa 3/metabolismo , Caspasa 3/genética , Peróxido de Hidrógeno/farmacología
6.
J Cell Mol Med ; 28(12): e18492, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38890795

RESUMEN

Intervertebral disc degeneration (IVDD) severely affects the work and the quality of life of people. We previously demonstrated that silencing activation transcription factor 3 (ATF3) blocked the IVDD pathological process by regulating nucleus pulposus cell (NPC) ferroptosis, apoptosis, inflammation, and extracellular matrix (ECM) metabolism. Nevertheless, whether miR-874-3p mediated the IVDD pathological process by targeting ATF3 remains unclear. We performed single-cell RNA sequencing (scRNA-seq) and bioinformatics analysis to identify ATF3 as a key ferroptosis gene in IVDD. Then, Western blotting, flow cytometry, ELISA, and animal experiments were performed to validate the roles and regulatory mechanisms of miR-874-3p/ATF3 signalling axis in IVDD. ATF3 was highly expressed in IVDD patients and multiple cell types of IVDD rat, as revealed by scRNA-seq and bioinformatics analysis. GO analysis unveiled the involvement of ATF3 in regulating cell apoptosis and ECM metabolism. Furthermore, we verified that miR-874-3p might protect against IVDD by inhibiting NPC ferroptosis, apoptosis, ECM degradation, and inflammatory response by targeting ATF3. In vivo experiments displayed the protective effect of miR-874-3p/ATF3 axis on IVDD. These findings propose the potential of miR-874-3p and ATF3 as biomarkers of IVDD and suggest that targeting the miR-874-3p/ATF3 axis may be a therapeutic target for IVDD.


Asunto(s)
Factor de Transcripción Activador 3 , Ferroptosis , Degeneración del Disco Intervertebral , MicroARNs , Núcleo Pulposo , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Degeneración del Disco Intervertebral/genética , Degeneración del Disco Intervertebral/metabolismo , Degeneración del Disco Intervertebral/patología , MicroARNs/genética , MicroARNs/metabolismo , Animales , Humanos , Ratas , Ferroptosis/genética , Masculino , Núcleo Pulposo/metabolismo , Núcleo Pulposo/patología , Análisis de la Célula Individual/métodos , Apoptosis/genética , Transducción de Señal , Femenino , Persona de Mediana Edad , Ratas Sprague-Dawley , Análisis de Secuencia de ARN/métodos , Matriz Extracelular/metabolismo , Adulto , Regulación de la Expresión Génica , Modelos Animales de Enfermedad , Biología Computacional/métodos
7.
Chem Biol Drug Des ; 103(6): e14565, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38862254

RESUMEN

Ferroptosis is a novel form of programmed cell death that is triggered by iron-dependent lipid peroxidation. Brusatol (BRU), a natural nuclear factor erythroid 2-related factor 2 inhibitor, exhibits potent anticancer effects in various types of cancer. However, the exact mechanism of BRU in the treatment of hepatocellular carcinoma (HCC) remains unknown. The anticancer effects of BRU in HCC were detected using cell counting kit-8 and colony formation assays and a xenograft model. RNA sequencing (RNA-seq) and bioinformatics analyses of HCC cells were utilized to elucidate the mechanism underlying the effects of BRU in HCC. The levels of reactive oxygen species (ROS), glutathione (GSH), malondialdehyde (MDA), and Fe2+ were measured using assay kits. The expression of activating transcription factor 3 (ATF3) was tested using RT-qPCR, western blotting, and immunofluorescence staining. The role of ATF3 in BRU-induced ferroptosis was examined using siATF3. BRU significantly inhibited HCC cell proliferation, both in vitro and in vivo. BRU activated the ferroptosis signaling pathway and increased ATF3 expression. Furthermore, ATF3 knockdown impeded BRU-induced ferroptosis. BRU suppressed HCC growth through ATF3-mediated ferroptosis, supporting BRU as a promising therapeutic agent for HCC.


Asunto(s)
Factor de Transcripción Activador 3 , Carcinoma Hepatocelular , Ferroptosis , Neoplasias Hepáticas , Cuassinas , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Ferroptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Animales , Cuassinas/farmacología , Cuassinas/química , Cuassinas/uso terapéutico , Línea Celular Tumoral , Ratones , Proliferación Celular/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , Transducción de Señal/efectos de los fármacos
8.
Cancer Res ; 84(15): 2450-2467, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38718305

RESUMEN

Peripheral T-cell lymphoma (PTCL) is a heterogeneous and aggressive disease with a poor prognosis. Histone deacetylase (HDAC) inhibitors have shown inhibitory effects on PTCL. A better understanding of the therapeutic mechanism underlying the effects of HDAC inhibitors could help improve treatment strategies. Herein, we found that high expression of HDAC3 is associated with poor prognosis in PTCL. HDAC3 inhibition suppressed lymphoma growth in immunocompetent mice but not in immunodeficient mice. HDAC3 deletion delayed the progression of lymphoma, reduced the lymphoma burden in the thymus, spleen, and lymph nodes, and prolonged the survival of mice bearing N-methyl-N-nitrosourea-induced lymphoma. Furthermore, inhibiting HDAC3 promoted the infiltration and enhanced the function of natural killer (NK) cells. Mechanistically, HDAC3 mediated ATF3 deacetylation, enhancing its transcriptional inhibitory activity. Targeting HDAC3 enhanced CXCL12 secretion through an ATF3-dependent pathway to stimulate NK-cell recruitment and activation. Finally, HDAC3 suppression improved the response of PTCL to conventional chemotherapy. Collectively, this study provides insights into the mechanism by which HDAC3 regulates ATF3 activity and CXCL12 secretion, leading to immune infiltration and lymphoma suppression. Combining HDAC3 inhibitors with chemotherapy may be a promising strategy for treating PTCL. Significance: Targeting HDAC3 suppresses progression of T-cell lymphoma by activating ATF3 to induce secretion of CXCL12 and promote infiltration of NK cells, providing an immunostimulatory approach for treating T-cell lymphoma patients.


Asunto(s)
Factor de Transcripción Activador 3 , Quimiocina CXCL12 , Inhibidores de Histona Desacetilasas , Histona Desacetilasas , Células Asesinas Naturales , Linfoma de Células T Periférico , Animales , Inhibidores de Histona Desacetilasas/farmacología , Ratones , Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética , Linfoma de Células T Periférico/patología , Linfoma de Células T Periférico/inmunología , Linfoma de Células T Periférico/metabolismo , Linfoma de Células T Periférico/tratamiento farmacológico , Linfoma de Células T Periférico/genética , Humanos , Quimiocina CXCL12/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Línea Celular Tumoral , Femenino , Masculino , Ratones Endogámicos C57BL , Pronóstico
9.
Stem Cells ; 42(8): 763-776, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38733123

RESUMEN

Endometrium fibrosis is the leading cause of uterine infertility. Macrophages participated in the occurrence and development of endometrial fibrosis. We previously reported that human umbilical cord multipotent stromal cells (hUC-MSCs) exerted their therapeutic effect in a macrophage-dependent manner in endometrial fibrosis. However precise mechanisms by which hUC-MSCs may influence macrophages in endometrial fibrosis remain largely unexplored. Here, we demonstrated that abnormal iron and lipid metabolism occurred in patients with intrauterine adhesions (IUA) and murine models. Ferroptosis has been proven to contribute to the progression of fibrotic diseases. Our results revealed that pharmacological activation of ferroptosis by Erastin aggravated endometrial fibrosis, while inhibition of ferroptosis by Ferrostatin-1 ameliorated endometrial fibrosis in vivo. Moreover, ferroptosis of macrophages was significantly upregulated in endometria of IUA murine models. Of note, transcriptome profiles revealed that CD36 gene expression was significantly increased in patients with IUA and immunofluorescence analysis showed CD36 protein was mainly located in macrophages. Silencing CD36 in macrophages could reverse cell ferroptosis. Dual luciferase reporter assay revealed that CD36 was the direct target of activation transcription factor 3 (ATF3). Furthermore, through establishing coculture system and IUA murine models, we found that hUC-MSCs had a protective role against macrophage ferroptosis and alleviated endometrial fibrosis related to decreased CD36 and ATF3. The effect of hUC-MSCs on macrophage ferroptosis was attributed to the upregulation of amphiregulin (AREG). Our data highlighted that macrophage ferroptosis occurred in endometrial fibrosis via the ATF3-CD36 pathway and hUC-MSCs protected against macrophage ferroptosis to alleviate endometrial fibrosis via secreting AREG. These findings provided a potential target for therapeutic implications of endometrial fibrosis.


Asunto(s)
Anfirregulina , Antígenos CD36 , Endometrio , Ferroptosis , Fibrosis , Macrófagos , Cordón Umbilical , Femenino , Humanos , Cordón Umbilical/citología , Cordón Umbilical/metabolismo , Animales , Macrófagos/metabolismo , Ratones , Anfirregulina/metabolismo , Anfirregulina/genética , Endometrio/metabolismo , Endometrio/patología , Antígenos CD36/metabolismo , Antígenos CD36/genética , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Células Madre Multipotentes/metabolismo
10.
BMC Musculoskelet Disord ; 25(1): 331, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38725009

RESUMEN

BACKGROUND: The development of neuropathic pain (NP) is one of the reasons why the pain is difficult to treat, and microglial activation plays an important role in NP. Recently, platelet-rich plasma (PRP) has emerged as a novel therapeutic method for knee osteoarthritis (KOA). However, it's unclarified whether PRP has analgesic effects on NP induced by KOA and the underlying mechanisms unknown. PURPOSE: To observe the analgesic effects of PRP on NP induced by KOA and explore the potential mechanisms of PRP in alleviating NP. METHODS: KOA was induced in male rats with intra-articular injections of monosodium iodoacetate (MIA) on day 0. The rats received PRP or NS (normal saline) treatment at days 15, 17, and 19 after modeling. The Von Frey and Hargreaves tests were applied to assess the pain-related behaviors at different time points. After euthanizing the rats with deep anesthesia at days 28 and 42, the corresponding tissues were taken for subsequent experiments. The expression of activating transcription factor 3 (ATF3) in dorsal root ganglia (DRG) and ionized-calcium-binding adapter molecule-1(Iba-1) in the spinal dorsal horn (SDH) was detected by immunohistochemical staining. In addition, the knee histological assessment was performed by hematoxylin-eosin (HE) staining. RESULTS: The results indicated that injection of MIA induced mechanical allodynia and thermal hyperalgesia, which could be reversed by PRP treatment. PRP downregulated the expression of ATF3 within the DRG and Iba-1 within the SDH. Furthermore, an inhibitory effect on cartilage degeneration was observed in the MIA + PRP group only on day 28. CONCLUSION: These results indicate that PRP intra-articular injection therapy may be a potential therapeutic agent for relieving NP induced by KOA. This effect could be attributed to downregulation of microglial activation and reduction in nerve injury.


Asunto(s)
Regulación hacia Abajo , Microglía , Neuralgia , Osteoartritis de la Rodilla , Plasma Rico en Plaquetas , Ratas Sprague-Dawley , Animales , Masculino , Neuralgia/terapia , Neuralgia/metabolismo , Microglía/metabolismo , Ratas , Osteoartritis de la Rodilla/terapia , Factor de Transcripción Activador 3/metabolismo , Ganglios Espinales/metabolismo , Modelos Animales de Enfermedad , Inyecciones Intraarticulares , Proteínas de Unión al Calcio/metabolismo , Ácido Yodoacético/toxicidad , Proteínas de Microfilamentos
11.
Artículo en Inglés | MEDLINE | ID: mdl-38777778

RESUMEN

BACKGROUND: Aristolochic acid nephropathy (AAN) is a rapidly progressive interstitial nephropathy caused by Aristolochic acid (AA). AAN is associated with the development of nephropathy and urothelial carcinoma. It is estimated that more than 100 million people worldwide are at risk of developing AAN. However, the underlying mechanisms driving renal deterioration in AAN remain poorly understood, and the treatment options are limited. METHODS: We obtained GSE27168 and GSE136276 series matrix data from the Gene Expression Omnibus (GEO) related to AAN. Using the R Studio environment, we applied the limma package and WGCNA package to identify co-differently expressed genes (co-DEGs). By GO/KEGG/GSVA analysis, we revealed common biological pathways. Subsequently, co-DEGs were subjected to the String database to construct a protein-protein interaction (PPI) network. The MCC algorithms implemented in the Cytohubba plugin were employed to identify hub genes. The hub genes were cross-referenced with the transcription factor (TF) database to identify hub TFs. Immune infiltration analysis was performed to identify key immune cell groups by utilizing CIBERSORT. The expressions of AAN-associated hub TFs were verified in vivo and in vitro. Finally, siRNA intervention was performed on the two TFs to verify their regulatory effect in AAN. RESULTS: Our analysis identified 88 co-DEGs through the "limma" and "WGCNA" R packages. A PPI network comprising 53 nodes and 34 edges was constructed with a confidence level >0.4. ATF3 and c-JUN were identified as hub TFs potentially linked to AAN. Additionally, expressions of ATF3 and c-JUN positively correlated with monocytes, basophils, and vessels, and negatively correlated with eosinophils and endothelial cells. We observed a significant increase in protein and mRNA levels of these two hub TFs. Furthermore, it was found that siRNA intervention targeting ATF3, but not c-JUN, alleviated cell damage induced by AA. The knockdown of ATF3 protects against oxidative stress and inflammation in the AAN cell model. CONCLUSION: This study provides novel insights into the role of ATF3 in AAN. The comprehensive analysis sheds light on the molecular mechanisms and identifies potential biomarkers and drug targets for AAN treatment.


Asunto(s)
Ácidos Aristolóquicos , Enfermedades Renales , Factores de Transcripción , Ácidos Aristolóquicos/toxicidad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Enfermedades Renales/inducido químicamente , Enfermedades Renales/genética , Animales , Ratones , Humanos , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Mapas de Interacción de Proteínas
12.
Cell Death Dis ; 15(5): 318, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710703

RESUMEN

Glioblastoma stem cells (GSCs) play a key role in glioblastoma (GBM) resistance to temozolomide (TMZ) chemotherapy. With the increase in research on the tumour microenvironment, exosomes secreted by GSCs have become a new focus in GBM research. However, the molecular mechanism by which GSCs affect drug resistance in GBM cells via exosomes remains unclear. Using bioinformatics analysis, we identified the specific expression of ABCB4 in GSCs. Subsequently, we established GSC cell lines and used ultracentrifugation to extract secreted exosomes. We conducted in vitro and in vivo investigations to validate the promoting effect of ABCB4 and ABCB4-containing exosomes on TMZ resistance. Finally, to identify the transcription factors regulating the transcription of ABCB4, we performed luciferase assays and chromatin immunoprecipitation-quantitative PCR. Our results indicated that ABCB4 is highly expressed in GSCs. Moreover, high expression of ABCB4 promoted the resistance of GSCs to TMZ. Our study found that GSCs can also transmit their highly expressed ABCB4 to differentiated glioma cells (DGCs) through exosomes, leading to high expression of ABCB4 in these cells and promoting their resistance to TMZ. Mechanistic studies have shown that the overexpression of ABCB4 in GSCs is mediated by the transcription factor ATF3. In conclusion, our results indicate that GSCs can confer resistance to TMZ in GBM by transmitting ABCB4, which is transcribed by ATF3, through exosomes. This mechanism may lead to drug resistance and recurrence of GBM. These findings contribute to a deeper understanding of the mechanisms underlying drug resistance in GBM and provide novel insights into its treatment.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP , Factor de Transcripción Activador 3 , Neoplasias Encefálicas , Resistencia a Antineoplásicos , Exosomas , Glioblastoma , Células Madre Neoplásicas , Temozolomida , Temozolomida/farmacología , Temozolomida/uso terapéutico , Glioblastoma/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Exosomas/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Línea Celular Tumoral , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Animales , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Ratones , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones Desnudos
13.
Environ Toxicol Pharmacol ; 108: 104469, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38759848

RESUMEN

We analyzed gene expression in THP-1 cells exposed to metal-based nanomaterials (NMs) [TiO2 (NM-100), ZnO (NM-110), SiO2 (NM-200), Ag (NM-300 K)]. A functional enrichment analysis of the significant differentially expressed genes (DEGs) identified the key modulated biological processes and pathways. DEGs were used to construct protein-protein interaction networks. NM-110 and NM-300 K induced changes in the expression of genes involved in oxidative and genotoxic stress, immune response, alterations of cell cycle, detoxification of metal ions and regulation of redox-sensitive pathways. Both NMs shared a number of highly connected protein nodes (hubs) including CXCL8, ATF3, HMOX1, and IL1B. NM-200 induced limited transcriptional changes, mostly related to the immune response; however, several hubs (CXCL8, ATF3) were identical with NM-110 and NM-300 K. No effects of NM-100 were observed. Overall, soluble nanomaterials NM-110 and NM-300 K exerted a wide variety of toxic effects, while insoluble NM-200 induced immunotoxicity; NM-100 caused no detectable changes on the gene expression level.


Asunto(s)
Mapas de Interacción de Proteínas , Plata , Titanio , Humanos , Titanio/toxicidad , Células THP-1 , Mapas de Interacción de Proteínas/efectos de los fármacos , Plata/toxicidad , Nanoestructuras/toxicidad , Nanopartículas del Metal/toxicidad , Óxido de Zinc/toxicidad , Óxido de Zinc/química , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Transcriptoma/efectos de los fármacos , Dióxido de Silicio/toxicidad , Interleucina-8/metabolismo , Interleucina-8/genética , Hemo-Oxigenasa 1
14.
Cell Death Differ ; 31(8): 1029-1043, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38762597

RESUMEN

Stress-adaptive mechanisms enabling cancer cells to survive under glucose deprivation remain elusive. N6-methyladenosine (m6A) modification plays important roles in determining cancer cell fate and cellular stress response to nutrient deficiency. However, whether m6A modification functions in the regulation of cancer cell survival under glucose deprivation is unknown. Here, we found that glucose deprivation reduced m6A modification levels. Increasing m6A modification resulted in increased hepatoma cell necrosis under glucose deprivation, whereas decreasing m6A modification had an opposite effect. Integrated m6A-seq and RNA-seq revealed potential targets of m6A modification under glucose deprivation, including the transcription factor FOSL1; further, glucose deprivation upregulated FOSL1 by inhibiting FOSL1 mRNA decay in an m6A-YTHDF2-dependent manner through reducing m6A modification in its exon1 and 5'-UTR regions. Functionally, FOSL1 protected hepatoma cells against glucose deprivation-induced necrosis in vitro and in vivo. Mechanistically, FOSL1 transcriptionally repressed ATF3 by binding to its promoter. Meanwhile, ATF3 and MAFF interacted via their leucine zipper domains to form a heterodimer, which competed with NRF2 for binding to antioxidant response elements in the promoters of NRF2 target genes, thereby inhibiting their transcription. Consequently, FOSL1 reduced the formation of the ATF3-MAFF heterodimer, thereby enhancing NRF2 transcriptional activity and the antioxidant capacity of glucose-deprived-hepatoma cells. Thus, FOSL1 alleviated the necrosis-inducing effect of glucose deprivation-induced reactive oxygen species accumulation. Collectively, our study uncovers the protective role of m6A-FOSL1-ATF3 axis in hepatoma cell necrosis under glucose deprivation, and may provide new targets for cancer therapy.


Asunto(s)
Carcinoma Hepatocelular , Glucosa , Neoplasias Hepáticas , Necrosis , Proteínas Proto-Oncogénicas c-fos , ARN Mensajero , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Glucosa/metabolismo , Glucosa/deficiencia , Humanos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Animales , Ratones , ARN Mensajero/metabolismo , ARN Mensajero/genética , Línea Celular Tumoral , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Ratones Desnudos
15.
Neurosci Lett ; 832: 137806, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38714229

RESUMEN

BACKGROUND: Trigeminal neuralgia (TN) is a common and difficult-to-treat neuropathic pain disorder in clinical practice. Previous studies have shown that Toll-like receptor 4 (TLR4) modulates the activation of the NF-κB pathway to affect neuropathic pain in rats. Voltage-gated sodium channels (VGSCs) are known to play an important role in neuropathic pain electrical activity. OBJECTIVE: To investigate whether TLR4 can regulate Nav1.3 through the TRAF6/NF-κB p65 pathway after infraorbital nerve chronic constriction injury (ION-CCI). STUDY DESIGN: ION-CCI modeling was performed on SD (Sprague Dawley) rats. To verify the success of the modeling, we need to detect the mechanical pain threshold and ATF3. Then, detecting the expression of TLR4, TRAF6, NF-κB p65, p-p65, and Nav1.3 in rat TG. Subsequently, investigate the role of TLR4/TRAF6/NF-κB pathway in ION-CCI model by intrathecal injections of LPS-rs (TLR4 antagonist), C25-140 (TRAF6 inhibitor), and PDTC (NF-κB p65 inhibitor). RESULTS: ION-CCI surgery decreased the mechanical pain threshold of rats and increased the expression of ATF3, TLR4, TRAF6, NF-κB p-p65 and Nav1.3, but there was no difference in NF-κB p65 expression. After inject antagonist or inhibitor of the TLR4/TRAF6/NF-κB pathway, the expression of Nav1.3 was decreased and mechanical pain threshold was increased. CONCLUSION: In the rat model of ION-CCI, TLR4 in the rat trigeminal ganglion regulates Nav1.3 through the TRAF6/NF-κB p65 pathway, and TLR4 antagonist alleviates neuropathic pain in ION-CCI rats.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.3 , Ratas Sprague-Dawley , Transducción de Señal , Factor 6 Asociado a Receptor de TNF , Receptor Toll-Like 4 , Animales , Receptor Toll-Like 4/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Masculino , Canal de Sodio Activado por Voltaje NAV1.3/metabolismo , Transducción de Señal/fisiología , FN-kappa B/metabolismo , Neuralgia del Trigémino/metabolismo , Ratas , Modelos Animales de Enfermedad , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción Activador 3/metabolismo , Umbral del Dolor/fisiología
16.
Cell Commun Signal ; 22(1): 240, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664711

RESUMEN

BACKGROUND: The repair of peripheral nerve injury poses a clinical challenge, necessitating further investigation into novel therapeutic approaches. In recent years, bone marrow mesenchymal stromal cell (MSC)-derived mitochondrial transfer has emerged as a promising therapy for cellular injury, with reported applications in central nerve injury. However, its potential therapeutic effect on peripheral nerve injury remains unclear. METHODS: We established a mouse sciatic nerve crush injury model. Mitochondria extracted from MSCs were intraneurally injected into the injured sciatic nerves. Axonal regeneration was observed through whole-mount nerve imaging. The dorsal root ganglions (DRGs) corresponding to the injured nerve were harvested to test the gene expression, reactive oxygen species (ROS) levels, as well as the degree and location of DNA double strand breaks (DSBs). RESULTS: The in vivo experiments showed that the mitochondrial injection therapy effectively promoted axon regeneration in injured sciatic nerves. Four days after injection of fluorescently labeled mitochondria into the injured nerves, fluorescently labeled mitochondria were detected in the corresponding DRGs. RNA-seq and qPCR results showed that the mitochondrial injection therapy enhanced the expression of Atf3 and other regeneration-associated genes in DRG neurons. Knocking down of Atf3 in DRGs by siRNA could diminish the therapeutic effect of mitochondrial injection. Subsequent experiments showed that mitochondrial injection therapy could increase the levels of ROS and DSBs in injury-associated DRG neurons, with this increase being correlated with Atf3 expression. ChIP and Co-IP experiments revealed an elevation of DSB levels within the transcription initiation region of the Atf3 gene following mitochondrial injection therapy, while also demonstrating a spatial proximity between mitochondria-induced DSBs and CTCF binding sites. CONCLUSION: These findings suggest that MSC-derived mitochondria injected into the injured nerves can be retrogradely transferred to DRG neuron somas via axoplasmic transport, and increase the DSBs at the transcription initiation regions of the Atf3 gene through ROS accumulation, which rapidly release the CTCF-mediated topological constraints on chromatin interactions. This process may enhance spatial interactions between the Atf3 promoter and enhancer, ultimately promoting Atf3 expression. The up-regulation of Atf3 induced by mitochondria further promotes the expression of downstream regeneration-associated genes and facilitates axon regeneration.


Asunto(s)
Factor de Transcripción Activador 3 , Axones , Roturas del ADN de Doble Cadena , Ganglios Espinales , Células Madre Mesenquimatosas , Mitocondrias , Regeneración Nerviosa , Especies Reactivas de Oxígeno , Nervio Ciático , Regulación hacia Arriba , Animales , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Mitocondrias/metabolismo , Mitocondrias/genética , Especies Reactivas de Oxígeno/metabolismo , Axones/metabolismo , Regeneración Nerviosa/genética , Regulación hacia Arriba/genética , Ratones , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Nervio Ciático/lesiones , Nervio Ciático/patología , Ganglios Espinales/metabolismo , Ratones Endogámicos C57BL , Masculino
17.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 125-129, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38650141

RESUMEN

Myocardial ischemia/reperfusion injury (MIRI) is an irreversible adverse event during the management of coronary heart disease that lacks effective controls. The underlying mechanism of MIRI still requires further investigation. Recent studies have suggested that overexpression of ATF3 protects against MIRI by regulating inflammatory responses, ferroptosis, and autophagy. The downstream target of ATF3, EGR1, also showed cardioprotective properties against MIRI by promoting autophagy. Therefore, further investigating the effect of ATF3/EGR1 pathway on MIRI-induced inflammation and autophagy is needed. Cardiomyocyte MIRI model was established by challenging H9C2 cells with hypoxia/reoxygenation (H/R). The ATF3 overexpression-H/R cell model by transfecting ATF3 plasmid into the H9C2 cell line. The transcription levels of ATF3 and EGR1 were determined using RT-qPCR, the levels of TNF-α and IL-6 were determined using ELISA kits, the protein expression of LC3 I, LC3 II, and P62 was determined via WB, and microstructure of H9C2 cell was observed by transmission electron microscopy (TEM). Overexpression of ATF3 significantly downregulated Egr1 levels, indicating that EGR1 might be the target of ATF3. By upregulating ATF3 levels, the extracellular levels of the inflammatory cytokines TNF-α and IL-6 significantly decreased, and the protein expression of the autophagy markers LC3 I, LC3 II, and P62 significantly increased. TEM results revealed that the cell line in the H/R-ATF3 group exhibited a higher abundance of autophagosome enclosures of mitochondria. The results indicated that ATF3/EGR1 may alleviate inflammation and improve autophagy in an H/R-induced MIRI model of cardiomyocytes.


Asunto(s)
Factor de Transcripción Activador 3 , Autofagia , Proteína 1 de la Respuesta de Crecimiento Precoz , Inflamación , Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Factor de Necrosis Tumoral alfa , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Autofagia/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Animales , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Ratas , Línea Celular , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Interleucina-6/metabolismo , Interleucina-6/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Transducción de Señal , Proteína Sequestosoma-1/metabolismo , Proteína Sequestosoma-1/genética
18.
Cell Death Dis ; 15(4): 290, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658567

RESUMEN

High-grade serous ovarian cancer (HGSOC) represents the most common and lethal subtype of ovarian cancer. Despite initial response to platinum-based standard therapy, patients commonly suffer from relapse that likely originates from drug-tolerant persister (DTP) cells. We generated isogenic clones of treatment-naïve and cisplatin-tolerant persister HGSOC cells. In addition, single-cell RNA sequencing of barcoded cells was performed in a xenograft model with HGSOC cell lines after platinum-based therapy. Published single-cell RNA-sequencing data from neo-adjuvant and non-treated HGSOC patients and patient data from TCGA were analyzed. DTP-derived cells exhibited morphological alterations and upregulation of epithelial-mesenchymal transition (EMT) markers. An aggressive subpopulation of DTP-derived cells showed high expression of the stress marker ATF3. Knockdown of ATF3 enhanced the sensitivity of aggressive DTP-derived cells to cisplatin-induced cell death, implying a role for ATF3 stress response in promoting a drug tolerant persister cell state. Furthermore, single cell lineage tracing to detect transcriptional changes in a HGSOC cell line-derived xenograft relapse model showed that cells derived from relapsed solid tumors express increased levels of EMT and multiple endoplasmic reticulum (ER) stress markers, including ATF3. Single cell RNA sequencing of epithelial cells from four HGSOC patients also identified a small cell population resembling DTP cells in all samples. Moreover, analysis of TCGA data from 259 HGSOC patients revealed a significant progression-free survival advantage for patients with low expression of the ATF3-associated partial EMT genes. These findings suggest that increased ATF3 expression together with partial EMT promote the development of aggressive DTP, and thereby relapse in HGSOC patients.


Asunto(s)
Factor de Transcripción Activador 3 , Cisplatino , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Neoplasias Ováricas , Humanos , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/genética , Femenino , Cisplatino/farmacología , Cisplatino/uso terapéutico , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Animales , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
19.
Circ Res ; 134(11): 1495-1511, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38686580

RESUMEN

BACKGROUND: Abdominal aortic aneurysm (AAA) is a catastrophic disease with little effective therapy, likely due to the limited understanding of the mechanisms underlying AAA development and progression. ATF3 (activating transcription factor 3) has been increasingly recognized as a key regulator of cardiovascular diseases. However, the role of ATF3 in AAA development and progression remains elusive. METHODS: Genome-wide RNA sequencing analysis was performed on the aorta isolated from saline or Ang II (angiotensin II)-induced AAA mice, and ATF3 was identified as the potential key gene for AAA development. To examine the role of ATF3 in AAA development, vascular smooth muscle cell-specific ATF3 knockdown or overexpressed mice by recombinant adeno-associated virus serotype 9 vectors carrying ATF3, or shRNA-ATF3 with SM22α (smooth muscle protein 22-α) promoter were used in Ang II-induced AAA mice. In human and murine vascular smooth muscle cells, gain or loss of function experiments were performed to investigate the role of ATF3 in vascular smooth muscle cell proliferation and apoptosis. RESULTS: In both Ang II-induced AAA mice and patients with AAA, the expression of ATF3 was reduced in aneurysm tissues but increased in aortic lesion tissues. The deficiency of ATF3 in vascular smooth muscle cell promoted AAA formation in Ang II-induced AAA mice. PDGFRB (platelet-derived growth factor receptor ß) was identified as the target of ATF3, which mediated vascular smooth muscle cell proliferation in response to TNF-alpha (tumor necrosis factor-α) at the early stage of AAA. ATF3 suppressed the mitochondria-dependent apoptosis at the advanced stage by upregulating its direct target BCL2. Our chromatin immunoprecipitation results also demonstrated that the recruitment of NFκB1 and P300/BAF/H3K27ac complex to the ATF3 promoter induces ATF3 transcription via enhancer activation. NFKB1 inhibitor (andrographolide) inhibits the expression of ATF3 by blocking the recruiters NFKB1 and ATF3-enhancer to the ATF3-promoter region, ultimately leading to AAA development. CONCLUSIONS: Our results demonstrate a previously unrecognized role of ATF3 in AAA development and progression, and ATF3 may serve as a novel therapeutic and prognostic marker for AAA.


Asunto(s)
Factor de Transcripción Activador 3 , Aneurisma de la Aorta Abdominal , Músculo Liso Vascular , Miocitos del Músculo Liso , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Animales , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/inducido químicamente , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Ratones , Masculino , Ratones Endogámicos C57BL , Apoptosis , Células Cultivadas , Angiotensina II , Proliferación Celular , Aorta Abdominal/patología , Aorta Abdominal/metabolismo , Modelos Animales de Enfermedad
20.
J Med Chem ; 67(8): 6810-6821, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38613772

RESUMEN

Anti-PD-L1 immunotherapy, a new lung cancer treatment, is limited to a few patients due to low PD-L1 expression and tumor immunosuppression. To address these challenges, the upregulation of PD-L1 has the potential to elevate the response rate and efficiency of anti-PD-L1 and alleviate the immunosuppression of the tumor microenvironment. Herein, we developed a novel usnic acid-derived Iridium(III) complex, Ir-UA, that boosts PD-L1 expression and converts "cold tumors" to "hot". Subsequently, we administered Ir-UA combined with anti-PD-L1 in mice, which effectively inhibited tumor growth and promoted CD4+ and CD8+ T cell infiltration. To our knowledge, Ir-UA is the first iridium-based complex to stimulate the expression of PD-L1 by explicitly regulating its transcription factors, which not only provides a promising platform for immune checkpoint blockade but, more importantly, provides an effective treatment strategy for patients with low PD-L1 expression.


Asunto(s)
Antígeno B7-H1 , Inmunoterapia , Iridio , Animales , Iridio/química , Iridio/farmacología , Antígeno B7-H1/metabolismo , Ratones , Humanos , Inmunoterapia/métodos , Factor de Transcripción Activador 3/metabolismo , Línea Celular Tumoral , Ratones Endogámicos C57BL , Microambiente Tumoral/efectos de los fármacos , Femenino , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antineoplásicos/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...