Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cancer Immunol Immunother ; 72(7): 2299-2308, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36884079

RESUMO

BACKGROUND: There is still no specific real-world data regarding the clinical activity of immune checkpoint inhibitors in the elderly with liver cancer. Our study aimed to compare the efficacy and safety of immune checkpoint inhibitors between patients aged ≥ 65 years and the younger group, while exploring their differences in genomic background and tumor microenvironment. METHODS: This retrospective study was conducted at two hospitals in China and included 540 patients treated with immune checkpoint inhibitors for primary liver cancer between January 2018 and December 2021. Patients' medical records were reviewed for clinical and radiological data and oncologic outcomes. The genomic and clinical data of patients with primary liver cancer were extracted and analyzed from TCGA-LIHC, GSE14520, and GSE140901 datasets. RESULTS: Ninety-two patients were classified as elderly and showed better progression-free survival (P = 0.027) and disease control rate (P = 0.014). No difference was observed in overall survival (P = 0.69) or objective response rate (P = 0.423) between the two age groups. No significant difference was reported concerning the number (P = 0.824) and severity (P = 0.421) of adverse events. The enrichment analyses indicated that the elderly group was linked to lower expression of oncogenic pathways, such as PI3K-Akt, Wnt, and IL-17. The elderly had a higher tumor mutation burden than younger patients. CONCLUSIONS: Our results indicated that immune checkpoint inhibitors might exhibit better efficacy in the elderly with primary liver cancer, with no increased adverse events. Differences in genomic characteristics and tumor mutation burden may partially explain these results.


Assuntos
Antineoplásicos Imunológicos , Neoplasias Hepáticas , Idoso , Humanos , Estudos Retrospectivos , Estudos de Coortes , Inibidores de Checkpoint Imunológico/efeitos adversos , Antineoplásicos Imunológicos/uso terapêutico , Fosfatidilinositol 3-Quinases , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Microambiente Tumoral
2.
Cancer Immunol Res ; 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38900485

RESUMO

Endoplasmic reticulum (ER) stress leads to hepatocellular carcinoma (HCC) progression. Small extracellular vesicles (sEVs) play a crucial role in modulating the tumor microenvironment (TME) by influencing cellular communication and immune responses. However, it is unclear whether ER stress modulates the TME through sEVs. In the current study, we investigated the effects and underlying mechanisms of ER stress on the HCC TME. In vivo and in vitro experiments showed that overactivated ER stress was a salient attribute of the immunosuppressive HCC TME. This was caused by the ATF4-promoted release of SNHG6-carrying sEVs, which attenuated T cell-mediated immune responses. Overall, SNHG6 modulated the immunosuppressive TME and aggravated ER stress. Meanwhile, targeting SNHG6 facilitated M1-like macrophage and CD8+ T-cell infiltration and decreased the proportion of M2-like macrophages. In addition, SNHG6 knockdown enhanced anti-PD-1 immunotherapeutic efficacy. Moreover, in HCC patients, overexpression of SNHG6 was associated with a lack of response to anti-PD-1 therapy and poor prognosis, whereas low SNHG6 expression was associated with improved therapeutic efficacy and prognoses. These data indicate that a correlation exists among ER stress, sEVs, an immunosuppressive HCC TME, and immunotherapeutic efficacy. Hence, SNHG6-targeted therapy may represent an effective strategy for patients with HCC.

3.
Front Immunol ; 14: 1333864, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38169837

RESUMO

Microbiome, including bacteria, fungi, and viruses, plays a crucial role in shaping distal and proximal anti-tumor immunity. Mounting evidence showed that commensal microbiome critically modulates immunophenotyping of hepatocellular carcinoma (HCC), a leading cause of cancer-related death. However, their role in anti-tumor surveillance of HCC is still poorly understood. Herein, we spotlighted growing interests in how the microbiome influences the progression and immunotherapeutic responses of HCC via changing local tumor microenvironment (TME) upon translocating to the sites of HCC through different "cell-type niches". Moreover, we summarized not only the associations but also the deep insight into the mechanisms of how the extrinsic microbiomes interplay with hosts to shape immune surveillance and regulate TME and immunotherapeutic responses. Collectively, we provided a rationale for a mechanism-guided fine-tuned microbiome to be neoadjuvant immunotherapy in the near future.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Microbiota , Humanos , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Vigilância Imunológica , Imunofenotipagem , Microambiente Tumoral
4.
Dis Markers ; 2023: 6729717, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36845017

RESUMO

Kinetochore-localized astrin- (SPAG5-) binding protein (KNSTRN) is mainly involved in mitosis. Somatic mutations in KNSTRN are known to lead to the occurrence and development of certain tumors. However, the role of KNSTRN in the tumor immune microenvironment (TIME) as a tumor prognostic biomarker and potential therapeutic target has not been clarified. Accordingly, in this study, we aimed to investigate the role of KNSTRN in the TIME. mRNA expression, cancer patient prognosis, and correlations between KNSTRN expression and immune component infiltration were analyzed using Genotype-Tissue Expression, The Cancer Genome Atlas, Cancer Cell Line Encyclopedia, Human Protein Atlas, ImmuCellAI, TIMER2.0, and KM-Plotter. The Genomics of Drug Sensitivity in Cancer database was used to evaluate the relationship between KNSTRN expression and the half maximal inhibitory concentration (IC50) of several anticancer drugs, and gene set variation analysis was performed. Data were visualized using R version 4.1.1. KNSTRN expression was upregulated in the majority of cancers and was associated with a worse prognosis. Additionally, KNSTRN expression was highly correlated with the infiltration of multiple immune components in the TIME and was related to a poor prognosis in tumor patients receiving immunotherapy. KNSTRN expression was also positively correlated with the IC50 of various anticancer drugs. In conclusion, KNSTRN may be a significant prognostic biomarker and promising target for oncotherapy in numerous cancers.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/genética , Proteínas de Ciclo Celular , Imunoterapia , Neoplasias/tratamento farmacológico , Neoplasias/genética , Prognóstico , Microambiente Tumoral
5.
Hepatol Int ; 17(6): 1444-1460, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37204655

RESUMO

BACKGROUND: Lowered nicotinamide adenine dinucleotide (NAD+) levels in tumor cells drive tumor hyperprogression during immunotherapy, and its restoration activates immune cells. However, the effect of lenvatinib, a first-line treatment for unresectable hepatocellular carcinoma (HCC), on NAD+ metabolism in HCC cells, and the metabolite crosstalk between HCC and immune cells after targeting NAD+ metabolism of HCC cells remain unelucidated. METHODS: Liquid chromatography-tandem mass spectrometry (LC-MS/MS) and ultra-high-performance liquid chromatography multiple reaction monitoring-mass spectrometry (UHPLC-MRM-MS) were used to detect and validate differential metabolites. RNA sequencing was used to explore mRNA expression in macrophages and HCC cells. HCC mouse models were used to validate the effects of lenvatinib on immune cells and NAD+ metabolism. The macrophage properties were elucidated using cell proliferation, apoptosis, and co-culture assays. In silico structural analysis and interaction assays were used to determine whether lenvatinib targets tet methylcytosine dioxygenase 2 (TET2). Flow cytometry was performed to assess changes in immune cells. RESULTS: Lenvatinib targeted TET2 to synthesize and increase NAD+ levels, thereby inhibiting decomposition in HCC cells. NAD+ salvage increased lenvatinib-induced apoptosis of HCC cells. Lenvatinib also induced CD8+ T cells and M1 macrophages infiltration in vivo. And lenvatinib suppressed niacinamide, 5-Hydroxy-L-tryptophan and quinoline secretion of HCC cells, and increased hypoxanthine secretion, which contributed to proliferation, migration and polarization function of macrophages. Consequently, lenvatinib targeted NAD+ metabolism and elevated HCC-derived hypoxanthine to enhance the macrophages polarization from M2 to M1. Glycosaminoglycan binding disorder and positive regulation of cytosolic calcium ion concentration were characteristic features of the reverse polarization. CONCLUSIONS: Targeting HCC cells NAD+ metabolism by lenvatinib-TET2 pathway drives metabolite crosstalk, leading to M2 macrophages reverse polarization, thereby suppressing HCC progression. Collectively, these novel insights highlight the role of lenvatinib or its combination therapies as promising therapeutic alternatives for HCC patients with low NAD+ levels or high TET2 levels.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Quinolinas , Animais , Camundongos , Humanos , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , NAD/metabolismo , NAD/farmacologia , NAD/uso terapêutico , Linfócitos T CD8-Positivos , Cromatografia Líquida , Linhagem Celular Tumoral , Espectrometria de Massas em Tandem , Macrófagos/metabolismo , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Hipoxantinas/metabolismo , Hipoxantinas/farmacologia , Hipoxantinas/uso terapêutico
6.
Eur J Med Res ; 28(1): 105, 2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36855207

RESUMO

BACKGROUND: Neonatal sepsis (NS), a life-threatening condition, is characterized by organ dysfunction and is the most common cause of neonatal death. However, the pathogenesis of NS is unclear and the clinical inflammatory markers currently used are not ideal for diagnosis of NS. Thus, exploring the link between immune responses in NS pathogenesis, elucidating the molecular mechanisms involved, and identifying potential therapeutic targets is of great significance in clinical practice. Herein, our study aimed to explore immune-related genes in NS and identify potential diagnostic biomarkers. Datasets for patients with NS and healthy controls were downloaded from the GEO database; GSE69686 and GSE25504 were used as the analysis and validation datasets, respectively. Differentially expressed genes (DEGs) were identified and Gene Set Enrichment Analysis (GSEA) was performed to determine their biological functions. Composition of immune cells was determined and immune-related genes (IRGs) between the two clusters were identified and their metabolic pathways were determined. Key genes with correlation coefficient > 0.5 and p < 0.05 were selected as screening biomarkers. Logistic regression models were constructed based on the selected biomarkers, and the diagnostic models were validated. RESULTS: Fifty-two DEGs were identified, and GSEA indicated involvement in acute inflammatory response, bacterial detection, and regulation of macrophage activation. Most infiltrating immune cells, including activated CD8 + T cells, were significantly different in patients with NS compared to the healthy controls. Fifty-four IRGs were identified, and GSEA indicated involvement in immune response and macrophage activation and regulation of T cell activation. Diagnostic models of DEGs containing five genes (PROS1, TDRD9, RETN, LOC728401, and METTL7B) and IRG with one gene (NSUN7) constructed using LASSO algorithm were validated using the GPL6947 and GPL13667 subset datasets, respectively. The IRG model outperformed the DEG model. Additionally, statistical analysis suggested that risk scores may be related to gestational age and birth weight, regardless of sex. CONCLUSIONS: We identified six IRGs as potential diagnostic biomarkers for NS and developed diagnostic models for NS. Our findings provide a new perspective for future research on NS pathogenesis.


Assuntos
Sepse Neonatal , Recém-Nascido , Humanos , Sepse Neonatal/diagnóstico , Sepse Neonatal/genética , Algoritmos , Biomarcadores , Inflamação , Modelos Logísticos
7.
Nan Fang Yi Ke Da Xue Xue Bao ; 40(10): 1480-1487, 2020 Oct 30.
Artigo em Chinês | MEDLINE | ID: mdl-33118501

RESUMO

OBJECTIVE: To observe the therapeutic effect of different doses of dihydroartemisinin (DHA) on atopic dermatitis (AD) in mice and explore the mechanism. METHODS: Forty-two C57BL/6 mice were randomly divided into 7 groups (n=6), including a blank control group, a 2, 4-dinitrochlorobenzene (DNCB)-induced AD model group, a solvent-treated group, 3 DHA treatment groups treated with 25, 75, and 125 mg/kg DHA, and a dexamethasone treatment group. The counts of skin scratches were recorded and the lesion scores were evaluated on a daily basis. After 7 consecutive days of treatment, skin tissues were sampled from the lesions on the back and ear of the mice for pathological examination with HE staining, Masson staining and toluidine blue staining. RESULTS: Treatment with 25, 75, and 125 mg/kg DHA and dexamethasone all alleviated AD symptoms of mice, reduced the severity scores of skin lesions, and ameliorated pathological changes of the skin tissue. DHA at 125 mg/kg produced the most obvious therapeutic effect and significantly alleviated mast cell infiltration in the lesions as compared with the other treatment groups (P < 0.05). CONCLUSIONS: DHA is effective for the treatment of AD in mice with an optimal dose of 125 mg/kg. The therapeutic effect of DHA is achieved probably through regulation of local immunity by inhibiting mast cell infiltration in the lesions.


Assuntos
Dermatite Atópica , Animais , Anti-Inflamatórios/uso terapêutico , Artemisininas , Citocinas , Dermatite Atópica/induzido quimicamente , Dermatite Atópica/tratamento farmacológico , Imunoglobulina E , Mastócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pele
8.
Toxicol Sci ; 173(2): 373-386, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31804693

RESUMO

Although studies have shown toxic effects of zinc oxide (ZnO) particles following inhalation, additional effects on injured lungs, which are characterized by dysfunction of the alveolar-capillary barriers, remain uncharacterized. To explore these additional effects, nano-sized ZnO (nZnO) and bulk-sized ZnO were applied to lipopolysaccharide (LPS)-challenged mouse lungs, which were used as a disease model of acute lung inflammation. An elevated Zn2+ concentration was detected in lung tissue after LPS plus nZnO exposure. Exposure to nZnO in LPS-challenged mice resulted in higher total cell number, proportion of neutrophils, and total protein level in bronchoalveolar lavage fluid. Intratracheal instillation of nZnO intensively aggravated LPS-induced lung inflammation that was accompanied by enhanced expression of interleukin-1ß, interleukin-6, monocyte chemotactic protein-1α, and granulocyte-macrophage colony stimulating factor. Catalase, glutathione, and total superoxide dismutase levels were significantly decreased, and the malondialdehyde level was obviously increased in the LPS plus nZnO group. 8-Hydroxyguanosine, a marker for DNA damage, was highly concentrated in the lungs from the LPS plus nZnO group. Furthermore, nZnO increased lung apoptosis in an acute lung inflammation model. Taken together, this evidence indicates that nZnO aggravates lung inflammation related to LPS. This enhancement effect may be mediated via oxidative stress, which can lead to DNA damage and apoptosis. This work is important because of the ever-increasing exposure of people to ZnO nanoparticles in industry. The identification of the toxic effects of nZnO and possible mechanisms revealed in this study provide valuable information for future studies.


Assuntos
Pulmão/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Pneumonia/induzido quimicamente , Traqueia/efeitos dos fármacos , Óxido de Zinco/toxicidade , Animais , Apoptose/efeitos dos fármacos , Quimiocinas/metabolismo , Dano ao DNA/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Fígado/patologia , Pulmão/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Estresse Oxidativo , Pneumonia/patologia , Traqueia/fisiopatologia , Óxido de Zinco/análise
9.
Am J Transl Res ; 11(1): 257-268, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30787984

RESUMO

Angiogenesis is an important step in skin wound repair. Angiogenesis is affected by the functions of many types of cells, especially endothelial cells. Cdc42 plays a vital role in endothelial cell function and vascular development; however, the role of Cdc42 in skin microvascular permeability and skin wound healing is unclear. This study investigated the involvement of Cdc42 in skin wound-healing processes based on its known roles in angiogenesis. Full-thickness skin wounds were created on wild-type and inducible vascular-endothelial-specific Cdc42-/- mice. Cdc42 deletion in endothelium affected wound healing in following ways. Reepithelialization of wounds in Cdc42-/- mice was delayed compared with that of wounds in wild-type mice. The degree of angiogenesis of wound granulation tissue was significantly lower in Cdc42-/- mice than in wild-type mice. Infiltration of F4/80+ macrophages and the expression of MCP-1, IL-1ß, and TNF-α were increased in the wound bed of Cdc42-/- mice compared with wild-type mice. These results confirm that Cdc42 in endothelium is required for angiogenesis and is an essential regulator of key skin wound-healing processes.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA