Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 623(7986): 432-441, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37914932

RESUMEN

Chromatin accessibility is essential in regulating gene expression and cellular identity, and alterations in accessibility have been implicated in driving cancer initiation, progression and metastasis1-4. Although the genetic contributions to oncogenic transitions have been investigated, epigenetic drivers remain less understood. Here we constructed a pan-cancer epigenetic and transcriptomic atlas using single-nucleus chromatin accessibility data (using single-nucleus assay for transposase-accessible chromatin) from 225 samples and matched single-cell or single-nucleus RNA-sequencing expression data from 206 samples. With over 1 million cells from each platform analysed through the enrichment of accessible chromatin regions, transcription factor motifs and regulons, we identified epigenetic drivers associated with cancer transitions. Some epigenetic drivers appeared in multiple cancers (for example, regulatory regions of ABCC1 and VEGFA; GATA6 and FOX-family motifs), whereas others were cancer specific (for example, regulatory regions of FGF19, ASAP2 and EN1, and the PBX3 motif). Among epigenetically altered pathways, TP53, hypoxia and TNF signalling were linked to cancer initiation, whereas oestrogen response, epithelial-mesenchymal transition and apical junction were tied to metastatic transition. Furthermore, we revealed a marked correlation between enhancer accessibility and gene expression and uncovered cooperation between epigenetic and genetic drivers. This atlas provides a foundation for further investigation of epigenetic dynamics in cancer transitions.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias , Humanos , Hipoxia de la Célula , Núcleo Celular , Cromatina/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Epigénesis Genética/genética , Transición Epitelial-Mesenquimal , Estrógenos/metabolismo , Perfilación de la Expresión Génica , Proteínas Activadoras de GTPasa/metabolismo , Metástasis de la Neoplasia , Neoplasias/clasificación , Neoplasias/genética , Neoplasias/patología , Secuencias Reguladoras de Ácidos Nucleicos/genética , Análisis de la Célula Individual , Factores de Transcripción/metabolismo
2.
Allergy ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727640

RESUMEN

BACKGROUND: Meteorin-like protein (METRNL)/Interleukin-41 (IL-41) is a novel immune-secreted cytokine/myokine involved in several inflammatory diseases. However, how METRNL exerts its regulatory properties on skin inflammation remains elusive. This study aims to elucidate the functionality and regulatory mechanism of METRNL in atopic dermatitis (AD). METHODS: METRNL levels were determined in skin and serum samples from patients with AD and subsequently verified in the vitamin D3 analogue MC903-induced AD-like mice model. The cellular target of METRNL activity was identified by multiplex immunostaining, single-cell RNA-seq and RNA-seq. RESULTS: METRNL was significantly upregulated in lesions and serum of patients with dermatitis compared to healthy controls (p <.05). Following repeated MC903 exposure, AD model mice displayed elevated levels of METRNL in both ears and serum. Administration of recombinant murine METRNL protein (rmMETRNL) ameliorated allergic skin inflammation and hallmarks of AD in mice, whereas blocking of METRNL signaling led to the opposite. METRNL enhanced ß-Catenin activation, limited the expression of Th2-related molecules that attract the accumulation of Arginase-1 (Arg1)hi macrophages, dendritic cells, and activated mast cells. CONCLUSIONS: METRNL can bind to KIT receptor and subsequently alleviate the allergic inflammation of AD by inhibiting the expansion of immune cells, and downregulating inflammatory gene expression by regulating the level of active WNT pathway molecule ß-Catenin.

3.
Gastroenterology ; 162(7): 2047-2062, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35271824

RESUMEN

BACKGROUND & AIMS: Checkpoint immunotherapy is largely ineffective in pancreatic ductal adenocarcinoma (PDAC). The innate immune nuclear factor (NF)-κB pathway promotes PDAC cell survival and stromal fibrosis, and is driven by Interleukin-1 Receptor Associated Kinase-4 (IRAK4), but its impact on tumor immunity has not been directly investigated. METHODS: We interrogated The Cancer Genome Atlas data to identify the correlation between NF-κB and T cell signature, and a PDAC tissue microarray (TMA) to correlate IRAK4 activity with CD8+ T cell abundance. We performed RNA sequencing (RNA-seq) on IRAK4-deleted PDAC cells, and single-cell RNA-seq on autochthonous KPC (p48-Cre/TP53f/f/LSL-KRASG12D) mice treated with an IRAK4 inhibitor. We generated conditional IRAK4-deleted KPC mice and complementarily used IRAK4 inhibitors to determine the impact of IRAK4 on T cell immunity. RESULTS: We found positive correlation between NF-κB activity, IRAK4 and T cell exhaustion from The Cancer Genome Atlas. We observed inverse correlation between phosphorylated IRAK4 and CD8+ T cell abundance in a PDAC tissue microarray. Loss of IRAK4 abrogates NF-κB activity, several immunosuppressive factors, checkpoint ligands, and hyaluronan synthase 2, all of which drive T cell dysfunction. Accordingly, conditional deletion or pharmacologic inhibition of IRAK4 markedly decreased tumor desmoplasia and increased the abundance and activity of infiltrative CD4+ and CD8+ T cells in KPC tumors. Single-cell RNA-seq showed myeloid and fibroblast reprogramming toward acute inflammatory responses following IRAK4 inhibition. These changes set the stage for successful combination of IRAK4 inhibitors with checkpoint immunotherapy, resulting in excellent tumor control and markedly prolonged survival of KPC mice. CONCLUSION: IRAK4 drives T cell dysfunction in PDAC and is a novel, promising immunotherapeutic target.


Asunto(s)
Carcinoma Ductal Pancreático , Quinasas Asociadas a Receptores de Interleucina-1 , Neoplasias Pancreáticas , Animales , Linfocitos T CD8-positivos/inmunología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Humanos , Inmunoterapia , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Ratones , FN-kappa B/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología
4.
Immunopharmacol Immunotoxicol ; 45(6): 692-700, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37358143

RESUMEN

OBJECTIVE: Treatment with TNF-α inhibitors improve psoriasis with minimize/minor neutrophils infiltration and CXCL-1/8 expression in psoriatic lesions. However, the fine mechanism of TNF-α initiating psoriatic inflammation by tuning keratinocytes is unclear. Our previous research identified the deficiency of intracellular galectin-3 was sufficient to promote psoriasis inflammation characterized by neutrophil accumulation. This study aims to investigate whether TNF-α participated in psoriasis development through dysregulating galectin-3 expression. METHODS: mRNA levels were assessed through quantitative real-time PCR. Flow cytometry was used to detect cell cycle/apoptosis. Western blot was used to evaluate the activation of the NF-κB signaling pathway. HE staining and immunochemistry were used to detect epidermal thickness and MPO expression, respectively. Specific small interfering RNA (siRNA) was used to knock down hsa-miR-27a-3p while plasmids transfection was used to overexpress galectin-3. Further, the multiMiR R package was utilized to predict microRNA-target interaction. RESULTS AND DISCUSSION: We found that TNF-α stimulation altered cell proliferation and differentiation and promoted the production of psoriasis-related inflammatory mediators along with the inhibition of galectin-3 expression in keratinocytes. Supplement of galectin-3 could counteract the rise of CXCL-1/8 but not the other phenotypes of keratinocytes induced by TNF-α. Mechanistically, inhibition of the NF-κB signaling pathway could counteract the decrease of galectin-3 and the increase of hsa-miR-27a-3p expression whereas silence of hsa-miR-27a-3p could counteract the decrease of galectin-3 expression induced by TNF-α treatment in keratinocytes. Intradermal injection of murine anti-CXCL-2 antibody greatly alleviated imiquimod-induced psoriasis-like dermatitis. CONCLUSION: TNF-α initiates psoriatic inflammation by increasing CXCL-1/8 in keratinocytes mediated by the axis of NF-κB-hsa-miR-27a-3p-galectin-3 pathway.


Asunto(s)
Galectina 3 , Queratinocitos , MicroARNs , Psoriasis , Factor de Necrosis Tumoral alfa , Factor de Necrosis Tumoral alfa/farmacología , Queratinocitos/metabolismo , Células HaCaT , Humanos , MicroARNs/genética , Quimiocina CXCL1/metabolismo , Interleucina-8/metabolismo , Galectina 3/genética , Psoriasis/genética , Psoriasis/patología , FN-kappa B/metabolismo , Transducción de Señal , Femenino , Animales , Ratones , Ratones Endogámicos C57BL
5.
Ann Allergy Asthma Immunol ; 129(3): 360-365.e1, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35598882

RESUMEN

BACKGROUND: Stevens-Johnson syndrome/toxic epidermal necrolysis (SJS/TEN) is a severe cutaneous adverse reaction to drugs with considerable morbidity and mortality. Immunomodulators for SJS/TEN including systemic corticosteroids and intravenous immunoglobulin (IVIG) have been widely used in clinical practice. Emerging evidence suggested the therapeutic effects of tumor necrosis factor-α antagonists on SJS/TEN. OBJECTIVE: To compare the efficacy and safety of IVIG and systemic steroids in conjunction with or without etanercept, a tumor necrosis factor-α inhibitor, for patients with SJS/TEN. METHODS: We undertook a retrospective review of 41 patients with SJS/TEN admitted to our institution from 2015 to February 2021. A total of 25 patients with integrated data were involved in this study, of which 14 patients were treated with IVIG and corticosteroids and 11 were in addition given etanercept. The clinical characteristics, duration of hospitalization, exposure time to high-dose steroids, and the total amount of systemic steroids were analyzed. RESULTS: In comparison to conventional therapy, conjunction with etanercept reduced the duration of hospitalization (13.5 vs 19.0 days; P = .01), the exposure time of high-dose steroids (7.1 vs 14.9 days; P = .01), and the overall amount of systemic steroid (925 mg vs 1412.5 mg; P = .03) in patients with SJS/TEN. No pronounced adverse effects were observed within 6 months of follow-up after the treatment. CONCLUSION: The add-in of etanercept at the time of initiating conventional therapy could be a superior option to accelerate disease recovery and reduce the high dose and total amount of systemic steroids without pronounced adverse events in patients with SJS/TEN.


Asunto(s)
Etanercept , Síndrome de Stevens-Johnson , Corticoesteroides/uso terapéutico , Etanercept/uso terapéutico , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Estudios Retrospectivos , Esteroides/uso terapéutico , Síndrome de Stevens-Johnson/tratamiento farmacológico , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico
6.
Gut ; 69(1): 122-132, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31076405

RESUMEN

OBJECTIVE: We investigated how pancreatic cancer developed resistance to focal adhesion kinase (FAK) inhibition over time. DESIGN: Pancreatic ductal adenocarcinoma (PDAC) tumours from KPC mice (p48-CRE; LSL-KRasG12D/wt; p53flox/wt) treated with FAK inhibitor were analysed for the activation of a compensatory survival pathway in resistant tumours. We identified pathways involved in the regulation of signal transducer and activator of transcription 3 (STAT3) signalling on FAK inhibition by gene set enrichment analysis and verified these outcomes by RNA interference studies. We also tested combinatorial approaches targeting FAK and STAT3 in syngeneic transplantable mouse models of PDAC and KPC mice. RESULTS: In KPC mice, the expression levels of phosphorylated STAT3 (pSTAT3) were increased in PDAC cells as they progressed on FAK inhibitor therapy. This progression corresponded to decreased collagen density, lowered numbers of SMA+ fibroblasts and downregulation of the transforming growth factor beta (TGF-ß)/SMAD signalling pathway in FAK inhibitor-treated PDAC tumours. Furthermore, TGF-ß production by fibroblasts in vitro drives repression of STAT3 signalling and enhanced responsiveness to FAK inhibitor therapy. Knockdown of SMAD3 in pancreatic cancer cells abolished the inhibitory effects of TGF-ß on pSTAT3. We further found that tumour-intrinsic STAT3 regulates the durability of the antiproliferative activity of FAK inhibitor, and combinatorial targeting of FAK and Janus kinase/STAT3 act synergistically to suppress pancreatic cancer progression in mouse models. CONCLUSION: Stromal depletion by FAK inhibitor therapy leads to eventual treatment resistance through the activation of STAT3 signalling. These data suggest that, similar to tumour-targeted therapies, resistance mechanisms to therapies targeting stromal desmoplasia may be critical to treatment durability.


Asunto(s)
Aminopiridinas/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Aminopiridinas/farmacología , Animales , Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/enzimología , Carcinoma Ductal Pancreático/patología , Colágeno/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Femenino , Fibroblastos/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Ratones Endogámicos , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína smad3/metabolismo , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Factor de Crecimiento Transformador beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Pharmacol Sci ; 140(1): 33-42, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31151763

RESUMEN

The present study aimed to investigate the functional components from Bushen Yijing Formula and their inhibition of endothelial-mesenchymal transition (EndMT) and fibrosis in human umbilical vascular endothelial cells (HUVECs). HUVEC fibrosis was induced by treatment of transforming growth factor ß (TGF-ß) as the cellular model. Expression of EndMT biomarker gene and cofactors were determined by quantitative real-time-PCR, western blotting, and immunofluorescence. Angiogenesis capacity of vein endothelial cells was evaluated using tube formation assay. Ursolic acid and drug-contained serum ameliorated EndMT biomarker gene expression changes and angiogenesis capacity suppression induced by TGF-ß treatment. Slug, Snail, and Twist gene expression and phosphorylation of mammalian target of rapamycin (mTOR) and AKT altered by TGF-ß in HUVECs were suppressed by ursolic acid and drug-contained serum. Treatment with the mTOR signaling pathway inhibitor, rapamycin, inhibited the phosphorylation of mTOR and AKT, decreased Snail and Vimentin protein levels, and increased VE-cad protein levels. Overexpression of Snail gene promoted expression of EndMT-related genes and suppressed angiogenesis in HUVECs, which were attenuated by application of ursolic acid and drug-contained serum. Ursolic acid from Bushen Yijing Formula inhibits human umbilical vein endothelial cell EndMT and fibrosis, mediated by AKT/mTOR signaling and Snail gene expression.


Asunto(s)
Medicamentos Herbarios Chinos/química , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Transcripción de la Familia Snail/genética , Factor de Crecimiento Transformador beta1/efectos adversos , Triterpenos/farmacología , Células Cultivadas , Fibrosis , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo , Triterpenos/aislamiento & purificación , Ácido Ursólico
8.
Int J Cancer ; 143(6): 1516-1529, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29607498

RESUMEN

Indoleamine 2,3-dioxygenase 1 (IDO1), known as IDO, catabolizes tryptophan through kynurenine pathway, whose activity is correlated with impaired clinical outcome of colorectal cancer. Here we showed that 1-L-MT, a canonical IDO inhibitor, suppressed proliferation of human colorectal cancer cells through inducing mitotic death. Our results showed that inhibition of IDO decreased the transcription of CDC20, which resulted in G2/M cycle arrest of HCT-116 and HT-29. Furthermore, 1-L-MT induced mitochondria injuries and caused apoptotic cancer cells. Importantly, 1-L-MT protected mice from azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colon carcinogenesis, with reduced mortality, tumor number and size. What is more, IDO1-/- mice exhibited fewer tumor burdens and reduced proliferation in the neoplastic epithelium, while, 1-L-MT did not exhibit any further protective effects on IDO-/- mice, confirming the critical role of IDO and the protective effect of 1-L-MT-mediated IDO inhibition in CRC. Furthermore, 1-L-MT also alleviated CRC in Rag1-/- mice, demonstrating the modulatory effects of IDO independent of its role in modulating adaptive immunity. Taken together, our findings validated that the anti-proliferation effect of 1-L-MT in vitro and the prevention of CRC in vivo were through IDO-induced cell cycle disaster of colon cancer cells. Our results identified 1-L-MT as a promising candidate for the chemoprevention of CRC.


Asunto(s)
Proteínas Cdc20/antagonistas & inhibidores , Colitis/complicaciones , Neoplasias del Colon/prevención & control , Inhibidores Enzimáticos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Mitosis , Triptófano/farmacología , Animales , Apoptosis , Azoximetano/toxicidad , Carcinógenos/toxicidad , Proteínas Cdc20/genética , Proteínas Cdc20/metabolismo , Ciclo Celular , Proliferación Celular , Colitis/inducido químicamente , Neoplasias del Colon/enzimología , Neoplasias del Colon/etiología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Proteínas de Homeodominio/fisiología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Triptófano/química , Triptófano/uso terapéutico , Células Tumorales Cultivadas
9.
Exp Cell Res ; 360(2): 320-327, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28942022

RESUMEN

NLRP3 inflammasome is a cytoplasmic multiprotein complex which plays a critical role in response to infection or injury, however, aberrant NLRP3 inflammasome activation is deleterious. In our study, we investigate the inhibitory effect of X-11-5-27, a daidzein derivative, on the NLRP3 inflammasome. The results showed that the activation of NLRP3 inflammasome was inhibited by X-11-5-27 in a dose-dependent manner, followed by a decrease in the cleavage of caspase-1 and maturation of IL-1ß. Furthermore, we found that X-11-5-27 significantly restrained the formation of NLRP3 inflammasome. At the same time, X-11-5-27 time- and dose-dependently decreased the production of ROS and superoxide. In addition, X-11-5-27 enhanced the activity of SOD to scavenge ROS release. This inhibitory effect of X-11-5-27 was due to the protection of mitochondrial homeostasis and was abolished after the treatment of rotenone. Notably, X-11-5-27 was found to trigger autophagy in macrophages, which in turn inhibited the NLRP3 inflammasome activation. Moreover, the phosphorylation states of the proteins in PI3K/AKT/mTOR signaling pathway were dramatically decreased after X-11-5-27 treatment. In conclusion, our results demonstrate that autophagy-mediated ROS reduction is responsible for X-11-5-27-induced NLRP3 flammasome inactivation. And these results may help guide decisions regarding the use of X-11-5-27 in relieving the inflammasome-driven hyper-inflammation.


Asunto(s)
Autofagia/efectos de los fármacos , Inflamasomas/antagonistas & inhibidores , Inflamasomas/efectos de los fármacos , Isoflavonas/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piperazinas/farmacología , Animales , Antiinflamatorios/farmacología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , Células Cultivadas , Inflamasomas/metabolismo , Inflamación/prevención & control , Isoflavonas/química , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
Int J Mol Sci ; 19(7)2018 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-29986455

RESUMEN

This study was conducted to envaluate whether glycine could alleviate Escherichia coli lipopolysaccharide (LPS)-induced intestinal injury by regulating intestinal epithelial energy status, protein synthesis, and inflammatory response via AMPK, mTOR, TLR4, and NOD signaling pathways. A total of 24 weanling piglets were randomly allotted to 1 of 4 treatments: (1) non-challenged control; (2) LPS-challenged control; (3) LPS + 1% glycine; (4) LPS + 2% glycine. After 28 days feeding, piglets were injected intraperitoneally with saline or LPS. The pigs were slaughtered and intestinal samples were collected at 4 h postinjection. The mRNA expression of key genes in these signaling pathways was measured by real-time PCR. The protein abundance was measured by Western blot analysis. Supplementation with glycine increased jejunal villus height/crypt depth ratio. Glycine also increased the jejunal and ileal protein content, RNA/DNA ratio, and jejunal protein/DNA ratio. The activities of citroyl synthetase in ileum, and α-ketoglutarate dehydrogenase complex in jejunum, were increased in the piglets fed diets supplemented with glycine. In addition, glycine decreased the jejunal and ileal phosphorylation of AMPKα, and increased ileal phosphorylation of mTOR. Furthermore, glycine downregulated the mRNA expression of key genes in inflammatory signaling. Meanwhile, glycine increased the mRNA expression of negative regulators of inflammatory signaling. These results indicate that glycine supplementation could improve energy status and protein synthesis by regulating AMPK and mTOR signaling pathways, and relieve inflammation by inhibiting of TLR4 and NOD signaling pathways to alleviate intestinal injury in LPS-challenged piglets.


Asunto(s)
Glicina/administración & dosificación , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Lipopolisacáridos/efectos adversos , Proteínas Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Receptor Toll-Like 4/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Escherichia coli , Glicina/farmacología , Íleon/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inyecciones Intraperitoneales , Yeyuno/metabolismo , Modelos Animales , Proteínas Quinasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Porcinos , Destete
11.
Cancer Discov ; 14(2): 208-210, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38327192

RESUMEN

SUMMARY: MHC-I downregulation is correlated with immunotherapy resistance in PDAC, but efficient strategies to increase cell-surface MHC-I are still lacking. This study by Sang, Zhou, Chen, Yu, and colleagues identified inhibition of tumor-intrinsic RIPK2 as a pharmacologic target to block the degradation of MHC-I on tumor cells and improved PDAC responses to anti-PD-1 immunotherapy. See related article by Sang et al., p. 326 (1) .


Asunto(s)
Inmunoterapia , Neoplasias Pancreáticas , Humanos , Vigilancia Inmunológica , Neoplasias Pancreáticas/tratamiento farmacológico
12.
Int Immunopharmacol ; 133: 112033, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38608446

RESUMEN

Psoriasis is an immuno-inflammatory disease characterized by excessive keratinocyte proliferation, requiring extensive lipids. 3-hydroxy-3-methylglutaryl-coenzyme A synthase 1 (HMGCS1) is an essential enzyme in the mevalonate pathway, involved in cholesterol synthesis and the inflammatory response. However, the role of HMGCS1 in psoriasis has remained elusive. This study aims to elucidate the mechanism by which HMGCS1 controls psoriasiform inflammation. We discovered an increased abundance of HMGCS1 in psoriatic lesions when analyzing two Gene Expression Omnibus (GEO) datasets and confirmed this in psoriatic animal models and psoriatic patients by immunohistochemistry. In a TNF-α stimulated psoriatic HaCaT cell line, HMGCS1 was found to be overexpressed. Knockdown of HMGCS1 using siRNA suppressed the migration and proliferation of HaCaT cells. Mechanistically, HMGCS1 downregulation also reduced the expression of IL-23 and the STAT3 phosphorylation level. In imiquimod-induced psoriatic mice, intradermal injection of HMGCS1 siRNA significantly decreased the expression of HMGCS1 in the epidermis, which in turn led to an improvement in the Psoriasis Area and Severity Index score, epidermal thickening, and pathological Baker score. Additionally, expression levels of inflammatory cytokines IL-23, IL1-ß, chemokine CXCL1, and innate immune mediator S100A7-9 were downregulated in the epidermis. In conclusion, HMGCS1 downregulation improved psoriasis in vitro and in vivo through the STAT3/IL-23 axis.


Asunto(s)
Hidroximetilglutaril-CoA Sintasa , Interleucina-23 , Queratinocitos , Psoriasis , Factor de Transcripción STAT3 , Animales , Femenino , Humanos , Masculino , Ratones , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células HaCaT , Hidroximetilglutaril-CoA Sintasa/metabolismo , Hidroximetilglutaril-CoA Sintasa/genética , Imiquimod , Interleucina-23/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Ratones Endogámicos BALB C , Psoriasis/inducido químicamente , Psoriasis/tratamiento farmacológico , Psoriasis/inmunología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética
13.
Cancer Discov ; 14(7): 1324-1355, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38683144

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) therapeutic resistance is largely attributed to a unique tumor microenvironment embedded with an abundance of cancer-associated fibroblasts (CAF). Distinct CAF populations were recently identified, but the phenotypic drivers and specific impact of CAF heterogeneity remain unclear. In this study, we identify a subpopulation of senescent myofibroblastic CAFs (SenCAF) in mouse and human PDAC. These SenCAFs are a phenotypically distinct subset of myofibroblastic CAFs that localize near tumor ducts and accumulate with PDAC progression. To assess the impact of endogenous SenCAFs in PDAC, we used an LSL-KRASG12D;p53flox;p48-CRE;INK-ATTAC (KPPC-IA) mouse model of spontaneous PDAC with inducible senescent cell depletion. Depletion of senescent stromal cells in genetic and pharmacologic PDAC models relieved immune suppression by macrophages, delayed tumor progression, and increased responsiveness to chemotherapy. Collectively, our findings demonstrate that SenCAFs promote PDAC progression and immune cell dysfunction. Significance: CAF heterogeneity in PDAC remains poorly understood. In this study, we identify a novel subpopulation of senescent CAFs that promotes PDAC progression and immunosuppression. Targeting CAF senescence in combination therapies could increase tumor vulnerability to chemo or immunotherapy. See related article by Ye et al., p. 1302.


Asunto(s)
Carcinoma Ductal Pancreático , Senescencia Celular , Miofibroblastos , Neoplasias Pancreáticas , Animales , Ratones , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Humanos , Miofibroblastos/metabolismo , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Microambiente Tumoral , Fibroblastos Asociados al Cáncer/metabolismo , Modelos Animales de Enfermedad
14.
EBioMedicine ; 90: 104507, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36893588

RESUMEN

BACKGROUND: Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease affecting multiple organs and tissues with high cellular heterogeneity. CD8+ T cell activity is involved in the SLE pathogenesis. However, the cellular heterogeneity and the underlying mechanisms of CD8+ T cells in SLE remain to be identified. METHODS: Single-cell RNA sequencing (scRNA-seq) of PBMCs from a SLE family pedigree (including 3 HCs and 2 SLE patients) was performed to identify the SLE-associated CD8+ T cell subsets. Flow cytometry analysis of a SLE cohort (including 23 HCs and 33 SLE patients), qPCR analysis of another SLE cohort (including 30 HCs and 25 SLE patients) and public scRNA-seq datasets of autoimmune diseases were employed to validate the finding. Whole-exome sequencing (WES) of this SLE family pedigree was used to investigate the genetic basis in dysregulation of CD8+ T cell subsets identified in this study. Co-culture experiments were performed to analyze the activity of CD8+ T cells. FINDINGS: We elucidated the cellular heterogeneity of SLE and identified a new highly cytotoxic CD8+ T cell subset, CD161-CD8+ TEMRA cell subpopulation, which was remarkably increased in SLE patients. Meanwhile, we discovered a close correlation between mutation of DTHD1 and the abnormal accumulation of CD161-CD8+ TEMRA cells in SLE. DTHD1 interacted with MYD88 to suppress its activity in T cells and DTHD1 mutation promoted MYD88-dependent pathway and subsequently increased the proliferation and cytotoxicity of CD161-CD8+ TEMRA cells. Furthermore, the differentially expressed genes in CD161-CD8+ TEMRA cells displayed a strong out-of-sample prediction for case-control status of SLE. INTERPRETATION: This study identified DTHD1-associated expansion of CD161-CD8+ TEMRA cell subpopulation is critical for SLE. Our study highlights genetic association and cellular heterogeneity of SLE pathogenesis and provides a mechanistical insight into the diagnosis and treatment of SLE. FUNDINGS: Stated in the Acknowledgements section of the manuscript.


Asunto(s)
Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Humanos , Linfocitos T CD8-positivos , Factor 88 de Diferenciación Mieloide/metabolismo , Subgrupos de Linfocitos T , Linfocitos T Citotóxicos/metabolismo , Lupus Eritematoso Sistémico/genética , Enfermedades Autoinmunes/metabolismo
15.
Cancer Cell ; 41(6): 1073-1090.e12, 2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-37236195

RESUMEN

Chronic activation of inflammatory pathways and suppressed interferon are hallmarks of immunosuppressive tumors. Previous studies have shown that CD11b integrin agonists could enhance anti-tumor immunity through myeloid reprograming, but the underlying mechanisms remain unclear. Herein we find that CD11b agonists alter tumor-associated macrophage (TAM) phenotypes by repressing NF-κB signaling and activating interferon gene expression simultaneously. Repression of NF-κB signaling involves degradation of p65 protein and is context independent. In contrast, CD11b agonism induces STING/STAT1 pathway-mediated interferon gene expression through FAK-mediated mitochondrial dysfunction, with the magnitude of induction dependent on the tumor microenvironment and amplified by cytotoxic therapies. Using tissues from phase I clinical studies, we demonstrate that GB1275 treatment activates STING and STAT1 signaling in TAMs in human tumors. These findings suggest potential mechanism-based therapeutic strategies for CD11b agonists and identify patient populations more likely to benefit.


Asunto(s)
Antígeno CD11b , Neoplasias , Humanos , Antígeno CD11b/agonistas , Inmunoterapia , Interferones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/inmunología , FN-kappa B/metabolismo , Transducción de Señal , Macrófagos Asociados a Tumores/inmunología
16.
J Exp Med ; 220(6)2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-36951731

RESUMEN

Tumor-associated macrophages (TAMs) are abundant in pancreatic ductal adenocarcinomas (PDACs). While TAMs are known to proliferate in cancer tissues, the impact of this on macrophage phenotype and disease progression is poorly understood. We showed that in PDAC, proliferation of TAMs could be driven by colony stimulating factor-1 (CSF1) produced by cancer-associated fibroblasts. CSF1 induced high levels of p21 in macrophages, which regulated both TAM proliferation and phenotype. TAMs in human and mouse PDACs with high levels of p21 had more inflammatory and immunosuppressive phenotypes. p21 expression in TAMs was induced by both stromal interaction and/or chemotherapy treatment. Finally, by modeling p21 expression levels in TAMs, we found that p21-driven macrophage immunosuppression in vivo drove tumor progression. Serendipitously, the same p21-driven pathways that drive tumor progression also drove response to CD40 agonist. These data suggest that stromal or therapy-induced regulation of cell cycle machinery can regulate both macrophage-mediated immune suppression and susceptibility to innate immunotherapy.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Ratones , Humanos , Neoplasias Pancreáticas/metabolismo , Macrófagos/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Inmunoterapia , Proliferación Celular , Microambiente Tumoral , Línea Celular Tumoral
17.
J Immunol Res ; 2022: 2787954, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36118416

RESUMEN

Cytokines like IL-17A have been consistently found to be elevated in psoriatic lesional skin, and therapeutic antibodies to IL-17 have demonstrated efficacy in treating psoriatic skin and joint disease. However, results about the circulating cytokines in psoriasis patients remained controversial. Anticytokine autoantibodies (ACAAs) were detected in various autoimmune diseases but remained largely unknown in psoriasis. We aimed to investigate the serum levels of cytokines and ACAAs in psoriasis patients. The study included 44 biologics-naive psoriasis patients and 40 healthy controls. Serum cytokines and the corresponding autoantibodies were measured by multiplex bead-based technology. The bioactivity of serum IL-17A was determined by IL-8 production in primary keratinocytes. Herein, we found serum levels of IL-12B (median: 6.16 vs. 9.03, p = 0.0194) and Th17 cytokines (IL-17A: median: 0.32 vs. 1.05, p = 0.0026; IL-22: median: 4.41 vs. 4.41, p = 0.0120) were increased in psoriasis patients. More interestingly, bioactive IL-17A was identified in a proportion of patients and positively correlated with disease severity. A few of cytokines were closely associated with each other and formed into a distinct panel in psoriasis. Of 13 anticytokine antibodies, anti-IL-22 was moderately lower (median: 262.8 vs.190.5, p = 0.0418), and anti-IL-15 was slightly higher (median: 25.5 vs. 30.5, p = 0.0069) in psoriasis than controls. None of ACAAs was related to disease severity. Consequently, the ratios of antibodies to cytokines varied with the pattern of cytokines. In summary, our finding suggested that the levels of circulating bioactive IL-17A were associated with disease activity in psoriasis patients. In contrast, the titers of ACAAs were not significantly altered nor correlated with disease severity. However, the functionality of ACAAs remains to be further demonstrated in vitro in future studies.


Asunto(s)
Productos Biológicos , Psoriasis , Anticuerpos Antiidiotipos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Autoanticuerpos , Productos Biológicos/uso terapéutico , Citocinas , Humanos , Interleucina-17 , Interleucina-8 , Psoriasis/tratamiento farmacológico
18.
Front Immunol ; 13: 817040, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35401573

RESUMEN

Microabscess of neutrophils in epidermis is one of the histological hallmarks of psoriasis. The axis of neutrophil-keratinocyte has been thought to play a critical role in the pathogenesis of psoriasis. However, the features and mechanism of interaction between the two cell types remain largely unknown. Herein, we found that blood neutrophils were increased in psoriasis patients, positively correlated with disease severity and highly expressed CD66b, but not CD11b and CD62L compared to healthy controls. Keratinocytes expressed high levels of psoriasis-related inflammatory mediators by direct and indirect interaction with neutrophils isolated from psoriasis patients and healthy controls. The capacity of neutrophils in provoking keratinocytes inflammatory response was comparable between the two groups and is dependent on IL-17A produced by itself. Neutrophils isolated from psoriasis patients displayed more transcriptome changes related to integrin and increased migration capacity toward keratinocytes with high CD11b expression on cell surface. Of interest, neutrophils were more susceptible to keratinocyte stimulation than to fibroblasts and human umbilical vein endothelial cells (HUVECs) in terms of CD11b expression and the production of ROS and NETs. In conclusion, neutrophils from psoriasis patients gain a strong capacity of IL-17A production and integrins expression that possibly facilitates their abilities to promote production of psoriasis-related inflammatory mediators and migration, a phenomenon likely induced by their interaction with keratinocytes but not with fibroblasts. These findings provide a proof-of-concept that development of new drugs targeting migration of neutrophils could be a more specific and safe solution to treat psoriasis.


Asunto(s)
Neutrófilos , Psoriasis , Células Endoteliales/metabolismo , Epidermis/patología , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-17/metabolismo , Queratinocitos/metabolismo , Neutrófilos/metabolismo , Psoriasis/patología
19.
Adv Rheumatol ; 62(1): 48, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36494762

RESUMEN

BACKGROUND: Systemic lupus erythematosus (SLE) is a systemic autoimmune disease that associates with aberrant activation of B lymphocytes and excessive autoantibodies. Interleukin 10 (IL-10)/interleukin 35 (IL-35) and IL-10/IL-35-producing regulatory B cells have been demonstrated to possess immunosuppressive functions during systemic lupus erythematosus. Here, we detected the proportion of CD19+CD24highCD27+ B cells as well as IL-10 and IL-35 levels in peripheral blood of SLE patients and healthy individuals, and investigated their relations with clinical features of SLE. METHODS: 41 SLE patients and 25 healthy controls were recruited. The patients were divided into groups based on SLEDAI score, anti-dsDNA antibody, rash, nephritis and hematological disorder. Flow cytometry was used to detect the proportion of CD24hiCD27+ B cells. ELISA was used to detect serum levels of IL-10 and IL-35. RESULTS: Our results showed that the CD19+CD24highCD27+ B population was decreased in active SLE patients, and anti-correlated with the disease activity. Of note, we found significant increase of IL-10 and decrease of IL-35 in SLE patients with disease activity score > 4, lupus nephritis or hematological disorders compared to those without related clinical features. CONCLUSIONS: Reduced CD19+CD24highCD27+ B cells expression may be involved in the pathogenesis of SLE. Moreover, we supposed that IL-35 instead of IL-10 played a crucial role in immune regulation during SLE disease.


Asunto(s)
Lupus Eritematoso Sistémico , Nefritis Lúpica , Humanos , Interleucina-10 , Antígenos CD19/metabolismo , Linfocitos B/metabolismo , Biomarcadores , Antígeno CD24/metabolismo
20.
Cancer Discov ; 12(12): 2774-2799, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36165893

RESUMEN

The effects of radiotherapy (RT) on tumor immunity in pancreatic ductal adenocarcinoma (PDAC) are not well understood. To better understand if RT can prime antigen-specific T-cell responses, we analyzed human PDAC tissues and mouse models. In both settings, there was little evidence of RT-induced T-cell priming. Using in vitro systems, we found that tumor-stromal components, including fibroblasts and collagen, cooperate to blunt RT efficacy and impair RT-induced interferon signaling. Focal adhesion kinase (FAK) inhibition rescued RT efficacy in vitro and in vivo, leading to tumor regression, T-cell priming, and enhanced long-term survival in PDAC mouse models. Based on these data, we initiated a clinical trial of defactinib in combination with stereotactic body RT in patients with PDAC (NCT04331041). Analysis of PDAC tissues from these patients showed stromal reprogramming mirroring our findings in genetically engineered mouse models. Finally, the addition of checkpoint immunotherapy to RT and FAK inhibition in animal models led to complete tumor regression and long-term survival. SIGNIFICANCE: Checkpoint immunotherapeutics have not been effective in PDAC, even when combined with RT. One possible explanation is that RT fails to prime T-cell responses in PDAC. Here, we show that FAK inhibition allows RT to prime tumor immunity and unlock responsiveness to checkpoint immunotherapy. This article is highlighted in the In This Issue feature, p. 2711.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Ratones , Animales , Humanos , Proteína-Tirosina Quinasas de Adhesión Focal , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/radioterapia , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Inmunoterapia , Microambiente Tumoral , Línea Celular Tumoral , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA