Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.451
Filtrer
1.
Mol Biol Rep ; 51(1): 988, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39285089

RÉSUMÉ

BACKGROUND: Nod2 is involved in innate immune responses to bacteria, regulation of metabolism, and sensitivity to cancer. A Nod2 polymorphism is associated with breast cancer, but the role of Nod2 in the development and progression of breast cancer is unknown. METHODS: Here, we tested the hypothesis that Nod2 protects mice from breast cancer using the 4T1 orthotopic model of mammary tumorigenesis. WT and Nod2-/- mice were injected with 4T1 mammary carcinoma cells and the development of tumors was monitored. A detailed analysis of the tumor transcriptome was performed and genes that were differentially expressed and pathways that were predicted to be altered between WT and Nod2-/- mice were identified. The activation of key signaling molecules involved in metabolism and development of cancer was studied. RESULTS: Our data demonstrate that Nod2-/- mice had a higher incidence and larger tumors than WT mice. Nod2-/- mice had increased expression of genes that promote DNA replication and cell division, and decreased expression of genes required for lipolysis, lipogenesis, and steroid biosynthesis compared with WT mice. Nod2-/- mice also had lower expression of genes required for adipogenesis and reduced levels of lipids compared with WT mice. The tumors in Nod2-/- mice had decreased expression of genes associated with PPARα/γ signaling, increased activation of STAT3, decreased activation of STAT5, and no change in the activation of ERK compared with WT mice. CONCLUSIONS: We conclude that Nod2 protects mice from the 4T1 orthotopic breast tumor, and that tumors in Nod2-/- mice are predicted to have increased DNA replication and cell proliferation and decreased lipid metabolism compared with WT mice.


Sujet(s)
Tumeurs du sein , Immunité innée , Protéine adaptatrice de signalisation NOD2 , Animaux , Protéine adaptatrice de signalisation NOD2/génétique , Protéine adaptatrice de signalisation NOD2/métabolisme , Souris , Femelle , Immunité innée/génétique , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Souris knockout , Lignée cellulaire tumorale , Transduction du signal/génétique , Régulation de l'expression des gènes tumoraux , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Souris de lignée BALB C , Transcriptome/génétique , Analyse de profil d'expression de gènes/méthodes
2.
Medicine (Baltimore) ; 103(37): e39511, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39287311

RÉSUMÉ

Breast cancer (BC) remains one of the most pervasive and complex malignancies. PANoptosis represents a recently identified cellular mechanism leading to programmed cell death. However, the prognostic implications and influence on the immune microenvironment of BC pertaining to PANoptosis-related genes (PRGs) remain significantly understudied. We conducted differential expression analysis to identify prognostic-Related PRGs by the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. Next, we identified the PANoptosis-related molecular subtype using the consensus clustering analysis, and constructed and validated the PANoptosis-related prognostic signature using LASSO and Cox regression analyses. ROC curves were employed to assess the performance of the signatures. Furthermore, drug sensitivity between low- and high-risk group were analysis. Finally, we conducted RT-qPCR to assess the gene expression levels involved in this signature. We categorized BC patients into 2 distinct molecular clusters based on PRGs and identified differentially expressed genes associated with prognosis. Subsequently, BC patients were then divided into 2 gene clusters. The identified PRGs molecular clusters and gene clusters demonstrated association with patient survival, immune system functions, and biological processes and pathways of BC. A prognostic signature comprising 5 genes was established, and BC patients were classified into low- and high-risk groups based on the risk scores. The ROC curves demonstrated that those in the low-risk category exhibited notably extended survival compared to the high-risk group. A nomogram model for patient survival was constructed based on the risk score in conjunction with other clinical features. High-risk group had higher tumor burden mutation, CSC index and lower StomalScore, ImmuneScore, and ESTIMATEScore. Subsequently, we established a correlation between the risk score and drug sensitivity among BC patients. Finally, qRT-PCR results showed that the expression of CXCL1, PIGR, and TNFRSF14 significantly decreased, while CXCL13 and NKAIN were significantly increased in BC tissues. We have developed a molecular clustering and prognostic signature based on PANoptosis to improve the prediction of BC prognosis. This discovery has the potential to not only assist in assessing overall patient prognosis but also to deepen our understanding of the underlying mechanisms of PANoptosis in BC pathogenesis.


Sujet(s)
Tumeurs du sein , Humains , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/mortalité , Femelle , Pronostic , Analyse de regroupements , Marqueurs biologiques tumoraux/génétique , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Courbe ROC , Adulte d'âge moyen
3.
Sci Rep ; 14(1): 21907, 2024 09 19.
Article de Anglais | MEDLINE | ID: mdl-39300166

RÉSUMÉ

Integrating immunotherapy with natural compounds holds promise in enhancing the immune system's ability to eliminate cancer cells. Cordyceps militaris, a traditional Chinese medicine, emerges as a promising candidate in this regard. This study investigates the effects of cordycepin and C. militaris ethanolic extract (Cm-EE) on sensitizing cancer cells and regulating immune responses against breast cancer (BC) and hepatocellular carcinoma (HCC) cells. Cordycepin, pentostatin and adenosine were identified in Cm-EE. Cordycepin treatment decreased HLA-ABC-positive cells in pre-treated cancer cells, while Cm-EE increased NKG2D ligand and death receptor expression. Additionally, cordycepin enhanced NKG2D receptor and death ligand expression on CD3-negative effector immune cells, particularly on natural killer (NK) cells, while Cm-EE pre-treatment stimulated IL-2, IL-6, and IL-10 production. Co-culturing cancer cells with effector immune cells during cordycepin or Cm-EE incubation resulted in elevated cancer cell death. These findings highlight the potential of cordycepin and Cm-EE in improving the efficacy of cancer immunotherapy for BC and HCC.


Sujet(s)
Cordyceps , Désoxyadénosine , Immunothérapie , Humains , Désoxyadénosine/pharmacologie , Cordyceps/composition chimique , Immunothérapie/méthodes , Lignée cellulaire tumorale , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/thérapie , Femelle , Tumeurs du foie/immunologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du sein/immunologie , Tumeurs du sein/traitement médicamenteux
4.
BMC Cancer ; 24(1): 1122, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39251967

RÉSUMÉ

BACKGROUND: Breast cancer (BRCA) remains to be among the main causes of cancer-associated mortality in women globally. HGH1 homolog (HGH1) has been reported to be associated with tumor immunity. However, the function of HGH1 in BRCA remains unclear. Therefore, the present study examined the potential role of HGH1 in BRCA. METHODS: The Cancer Genome Atlas (TCGA) databases and Gene Expression Omnibus (GEO) were used to obtain RNA-seq data for BRCA. A protein localization of HGH1 was determined by using the Human Protein Atlas (HPA), and immunohistochemistry (IHC) staining revealed an upregulation in the expression of HGH1 in clinical BRCA tissues. Xenograft mice were used to test tumor growth and HGH1 expression in breast cancer cells. The protein interaction information of HGH1 was analyzed using the GeneMANIA website. Based on univariate Cox regression and Kaplan-Meier methods, we evaluated the role of HGH1 in BRCA prognosis. HGH1-related differentially expressed genes were analyzed using GO, KEGG, and GSEA. We also examined the relationship between HGH1 expression, immune checkpoints, and immune infiltration. CCK-8, EdU, and colony formation assays were used to measure cell proliferation, and western blot analysis was used to evaluate HGH1's role in BRCA. RESULTS: IHC results showed that the expression of HGH1 was significantly upregulated in BRCA tissues compared to normal tissues. High levels of HGH1 expression was associated with worse clinical features and a worse prognosis. HGH1 expression was an independent predictor of BRCA outcomes in both univariate and multivariate analyses. Functionally, western blot analysis showed that HGH1 is implicated in cell cycle. As well, knocking down HGH1 significantly reduced BRCA cells' proliferative abilities. Crucially, HGH1 expression levels were positively correlated with Th2 cell infiltration and negatively correlated with Tcm cell infiltration. CONCLUSION: Biomarkers such as HGH1 can reliably predict prognosis in BRCA patients.


Sujet(s)
Tumeurs du sein , Cycle cellulaire , Humains , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/mortalité , Femelle , Pronostic , Animaux , Souris , Cycle cellulaire/génétique , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale
5.
Mol Cancer ; 23(1): 187, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39242519

RÉSUMÉ

BACKGROUND: The plasma concentrations of acyl coenzyme A binding protein (ACBP, also known as diazepam-binding inhibitor, DBI, or 'endozepine') increase with age and obesity, two parameters that are also amongst the most important risk factors for cancer. METHODS: We measured ACBP/DBI in the plasma from cancer-free individuals, high-risk patients like the carriers of TP53 or BRCA1/2 mutations, and non-syndromic healthy subjects who later developed cancer. In mice, the neutralization of ACBP/DBI was used in models of non-small cell lung cancer (NSCLC) and breast cancer development and as a combination treatment with chemoimmunotherapy (chemotherapy + PD-1 blockade) in the context of NSCLC and sarcomas. The anticancer T cell response upon ACBP/DBI neutralization was characterized by flow cytometry and single-cell RNA sequencing. RESULTS: Circulating levels of ACBP/DBI were higher in patients with genetic cancer predisposition (BRCA1/2 or TP53 germline mutations) than in matched controls. In non-syndromic cases, high ACBP/DBI levels were predictive of future cancer development, and especially elevated in patients who later developed lung cancer. In preclinical models, ACBP/DBI neutralization slowed down breast cancer and NSCLC development and enhanced the efficacy of chemoimmunotherapy in NSCLC and sarcoma models. When combined with chemoimmunotherapy, the neutralizing monoclonal antibody against ACBP/DBI reduced the frequency of regulatory T cells in the tumor bed, modulated the immune checkpoint profile, and increased activation markers. CONCLUSION: These findings suggest that ACBP/DBI acts as an endogenous immune suppressor. We conclude that elevation of ACBP/DBI constitutes a risk factor for the development of cancer and that ACBP/DBI is an actionable target for improving cancer immunosurveillance.


Sujet(s)
Marqueurs biologiques tumoraux , Animaux , Femelle , Humains , Souris , Tumeurs du sein/immunologie , Tumeurs du sein/diagnostic , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/diagnostic , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Lignée cellulaire tumorale , Surveillance immunologique , Tumeurs du poumon/immunologie , Tumeurs du poumon/diagnostic , Tumeurs du poumon/métabolisme , Tumeurs/diagnostic , Tumeurs/immunologie , Tumeurs/étiologie , Facteurs de risque
6.
Technol Cancer Res Treat ; 23: 15330338241281285, 2024.
Article de Anglais | MEDLINE | ID: mdl-39248214

RÉSUMÉ

Objectives: Heparanase (HPSE), an endoglycosidase that cleaves heparan sulfate, regulates various biological processes related to tumor progression. We explore the prognostic value of HPSE and its relationship with immunotherapy response in patients with breast cancer, to improve the effectiveness of immunotherapy and increase the survival outcomes. Methods: In the study, we explored the prognostic value of HPSE through the The Cancer Genome Atlas (TCGA) database. By using the single-sample gene set enrichment analysis (ssGSEA) method, we measured the infiltration levels of 24 immune cell types in the tumor microenvironment. Cancer Therapeutics Response Portal (CTRP) and PRISM datasets provide the area under the dose-response curve (AUC) to measure drug sensitivity. Using nomograms, we predicted overall survival ability. In vivo studies, we investigated the relationship between HPSE and immune checkpoint proteins and pro-inflammatory cytokines by immunohistochemistry of Triple-Negative Breast Cancer tumors in mice. Results: Our model demonstrated that the integrating of HPSE with the clinical stage effectively predicts patients' survival time, highlighting high HPSE expression as a prognostic risk factor for breast cancer. Then the Receiver Operating Characteristic (ROC) curve [AUC of 1 year = 0.747, AUC of 3 years = 0.731] and Decision Curve Analysis (DCA) curve illustrated the satisfactory discriminative capacity of our model, emphasizing its valuable clinical applicability. Immune-related results showed that HPSE correlates strongly with immune infiltrating cells, immune-related genes, and the anti-cancer immunity cycle. In vivo studies have demonstrated that HPSE in breast cancer is associated with increased expression of immune checkpoint proteins CD274 and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and is positively correlated with the pro-inflammatory cytokine TNF-α. Meanwhile, we analyzed the 11 types of drugs that are sensitive to the HPSE gene. Conclusion: Our results show that HPSE can serve as an effective biomarker to predict the prognosis of breast cancer patients and reflect the impact of immunotherapy.


Sujet(s)
Antigène CD274 , Marqueurs biologiques tumoraux , Tumeurs du sein , Antigène CTLA-4 , Régulation de l'expression des gènes tumoraux , Glucuronidase , Microenvironnement tumoral , Humains , Femelle , Pronostic , Glucuronidase/métabolisme , Glucuronidase/génétique , Animaux , Souris , Marqueurs biologiques tumoraux/métabolisme , Antigène CTLA-4/métabolisme , Tumeurs du sein/mortalité , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/métabolisme , Microenvironnement tumoral/immunologie , Antigène CD274/métabolisme , Antigène CD274/génétique , Protéines de points de contrôle immunitaires/métabolisme , Protéines de points de contrôle immunitaires/génétique , Courbe ROC , Nomogrammes , Biologie informatique/méthodes , Analyse de profil d'expression de gènes , Bases de données génétiques
7.
Clin Exp Med ; 24(1): 230, 2024 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-39325106

RÉSUMÉ

The escalating incidence of breast cancer (BC) in women underscores its grave health threat. Current molecular insights into BC's post-adjuvant therapy cure remain elusive, necessitating active treatment explorations. Immunotherapy, notably chemotherapy-induced immunogenic cell death (ICD), has emerged as a promising BC therapy. ICD harnesses chemotherapeutics to activate anti-tumor immunity via DAMPs, fostering long-term T-cell memory and primary BC cure. Besides chemotherapy drugs, Nanodrugs, traditional Chinese medicine (TCM) and ICIs also induce ICD, boosting immune response. ICIs, like PD-1/PD-L1 inhibitors, revolutionize cancer treatment but face limited success in cold tumors. Thus, ICD induction combined with ICIs is studied extensively for BC immunotherapy. This article reviews the mechanism of ICD related drugs in BC and provides reference for the research and development of BC treatment, in order to explore more effective clinical treatment of BC, we hope to explore more ICD inducers and make ICIs more effective vaccines.


Sujet(s)
Tumeurs du sein , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/immunologie , Tumeurs du sein/thérapie , Femelle , Antinéoplasiques/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie/méthodes , Mort cellulaire/effets des médicaments et des substances chimiques
8.
Int J Mol Sci ; 25(17)2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39273217

RÉSUMÉ

Currently, therapy for early-stage human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC) is based on the combination of trastuzumab and pertuzumab plus chemotherapy in a neoadjuvant regimen. The INMUNOHER study aimed to detect immunological markers in peripheral blood and their association with treatment response. Sixty-two HER2+ BC patients were recruited. Pre-treatment samples were obtained before the start of treatment, while post-treatment samples were obtained after completing therapy and before surgery and were analyzed by flow cytometry. The pathologic complete response (pCR) rate achieved was 82.3%. The expression of the NKp30, PD-1, and TIM-3 receptors was reduced in the Natural Killer (NK)-CD56dim subset of patients who did not achieve pCR. Following therapy, many changes were found in leukocytes, including alterations in T cell lymphocyte proportions. Also, the percentage of NK cells decreased, and several phenotypic changes were observed in this population. After treatment, IFN-γ production by NK cells against HER2+-cells with or without trastuzumab was significantly reduced. HER2-targeted therapy plus chemotherapy demonstrated high efficacy in most patients, reducing the statistical power for finding immunological markers. However, NK subset phenotypes correlated better with response groups, and numerous changes in the percentage of leukocytes and T and NK cells, as well as changes in the functionality of NK cells, were observed in most patients after treatment, encouraging further research into these immune populations.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Cellules tueuses naturelles , Traitement néoadjuvant , Récepteur ErbB-2 , Trastuzumab , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/immunologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Trastuzumab/usage thérapeutique , Trastuzumab/administration et posologie , Femelle , Traitement néoadjuvant/méthodes , Récepteur ErbB-2/métabolisme , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie , Adulte d'âge moyen , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Sujet âgé
9.
Cells ; 13(18)2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39329702

RÉSUMÉ

The tumor microenvironment (TME) is crucial in cancer development and therapeutic response. Immunotherapy is increasingly recognized as a critical component of cancer treatment. While immunotherapies have shown efficacy in various cancers, including breast cancer, patient responses vary widely. Some patients receive significant benefits, while others experience minimal or no improvement. This disparity underscores the complexity and diversity of the immune system. In this study, we investigated the immune landscape and cell-cell communication within the TME of breast cancer through integrated analysis of bulk and single-cell RNA sequencing data. We established profiles of tumor immune infiltration that span across a broad spectrum of adaptive and innate immune cells. Our clustering analysis of immune infiltration identified three distinct patient groups: high T cell abundance, moderate infiltration, and low infiltration. Patients with low immune infiltration exhibited the poorest survival rates, while those in the moderate infiltration group showed better outcomes than those with high T cell abundance. Moreover, the high cell abundance group was associated with a greater tumor burden and higher rates of TP53 mutations, whereas the moderate infiltration group was characterized by a lower tumor burden and elevated PIK3CA mutations. Analysis of an independent single-cell RNA-seq breast cancer dataset confirmed the presence of similar infiltration patterns. Further investigation into ligand-receptor interactions within the TME unveiled significant variations in cell-cell communication patterns among these groups. Notably, we found that the signaling pathways SPP1 and EGF were exclusively active in the low immune infiltration group, suggesting their involvement in immune suppression. This work comprehensively characterizes the composition and dynamic interplay in the breast cancer TME. Our findings reveal associations between the extent of immune infiltration and clinical outcomes, providing valuable prognostic information for patient stratification. The unique mutations and signaling pathways associated with different patient groups offer insights into the mechanisms underlying diverse tumor immune infiltration and the formation of an immunosuppressive tumor microenvironment.


Sujet(s)
Tumeurs du sein , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Femelle , Pronostic , Mutation/génétique , Analyse sur cellule unique , Lymphocytes TIL/immunologie
10.
Cells ; 13(18)2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39329710

RÉSUMÉ

Regulatory T cells (Tregs) play a key role in maintaining immune homeostasis and preventing autoimmunity through their immunosuppressive function. There have been numerous reports confirming that high levels of Tregs in the tumor microenvironment (TME) are associated with a poor prognosis, highlighting their role in promoting an immunosuppressive environment. In breast cancer (BC), Tregs interact with cancer cells, ultimately leading to the suppression of immune surveillance and promoting tumor progression. This review discusses the dual role of Tregs in breast cancer, and explores the controversies and therapeutic potential associated with targeting these cells. Researchers are investigating various strategies to deplete or inhibit Tregs, such as immune checkpoint inhibitors, cytokine antagonists, and metabolic inhibition. However, the heterogeneity of Tregs and the variable precision of treatments pose significant challenges. Understanding the functional diversity of Tregs and the latest advances in targeted therapies is critical for the development of effective therapies. This review highlights the latest approaches to Tregs for BC treatment that both attenuate Treg-mediated immunosuppression in tumors and maintain immune tolerance, and advocates precise combination therapy strategies to optimize breast cancer outcomes.


Sujet(s)
Tumeurs du sein , Lymphocytes T régulateurs , Microenvironnement tumoral , Humains , Tumeurs du sein/immunologie , Tumeurs du sein/thérapie , Tumeurs du sein/anatomopathologie , Lymphocytes T régulateurs/immunologie , Femelle , Microenvironnement tumoral/immunologie , Animaux , Immunothérapie/méthodes
11.
Sci Rep ; 14(1): 20403, 2024 09 02.
Article de Anglais | MEDLINE | ID: mdl-39223212

RÉSUMÉ

Leukemia inhibitory factor (LIF) is involved in the progression of different cancers. In this study, we investigated the effect of anti-LIF antibodies on immune-related gene expression in the Balb/c mouse model of breast cancer. To immunize mice against LIF, recombinant LIF with Freund adjuvant was injected into the test group, whereas the control group received phosphate-buffered saline with adjuvant. Tumor induction (4T1 cell line) was performed by increasing the antibody titer. The expression of immune-related genes was evaluated by real-time PCR. The anti-LIF titer was significantly increased in the immunized group. The expression of genes related to the differentiation of T helper (Th)-1, Th-2, and Th-17 cells was significantly higher in the immunized group than in the control group. In addition, anti-LIF did not have a significant effect on the expression of genes related to the differentiation of regulatory T cells, and immune checkpoint-associated genes. Additionally, the test group had higher survival and lower tumor development rates. The results demonstrated that the anti-LIF antibody may potentially play a role in the differentiation of immune cells or immune responses. However, further studies utilizing advanced techniques are necessary to validate its function.


Sujet(s)
Tumeurs du sein , Facteur inhibiteur de la leucémie , Souris de lignée BALB C , Animaux , Femelle , Facteur inhibiteur de la leucémie/génétique , Facteur inhibiteur de la leucémie/métabolisme , Facteur inhibiteur de la leucémie/immunologie , Souris , Tumeurs du sein/immunologie , Tumeurs du sein/génétique , Modèles animaux de maladie humaine , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Anticorps/immunologie
12.
Sci Rep ; 14(1): 20432, 2024 09 03.
Article de Anglais | MEDLINE | ID: mdl-39227722

RÉSUMÉ

Breast cancer (BC) stands as a predominant global malignancy, significantly contributing to female mortality. Recently uncovered, histone lysine lactylation (kla) has assumed a crucial role in cancer progression. However, the correlation with lncRNAs remains ambiguous. Scrutinizing lncRNAs associated with Kla not only improves clinical breast cancer management but also establishes a groundwork for antitumor drug development. We procured breast tissue samples, encompassing both normal and cancerous specimens, from The Cancer Genome Atlas (TCGA) database. Utilizing Cox regression and XGBoost methods, we developed a prognostic model using identified kla-related lncRNAs. The model's predictive efficacy underwent validation across training, testing, and the overall cohort. Functional analysis concerning kla-related lncRNAs ensued. We identified and screened 8 kla-related lncRNAs to formulate the risk model. Pathway analysis disclosed the connection between immune-related pathways and the risk model of kla-related lncRNAs. Significantly, the risk scores exhibited a correlation with both immune cell infiltration and immune function, indicating a clear association. Noteworthy is the observation that patients with elevated risk scores demonstrated an increased tumor mutation burden (TMB) and decreased tumor immune dysfunction and exclusion (TIDE) scores, suggesting heightened responses to immune checkpoint blockade. Our study uncovers a potential link between Kla-related lncRNAs and BC, providing innovative therapeutic guidelines for BC management.


Sujet(s)
Tumeurs du sein , Lysine , ARN long non codant , Microenvironnement tumoral , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Femelle , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Lysine/métabolisme , Pronostic , Marqueurs biologiques tumoraux/génétique , Régulation de l'expression des gènes tumoraux
13.
Front Immunol ; 15: 1444589, 2024.
Article de Anglais | MEDLINE | ID: mdl-39253073

RÉSUMÉ

Recent years have seen an outstanding growth in the understanding of connections between diet-induced obesity, dysbiosis and alterations in the tumor microenvironment. Now we appreciate that gut dysbiosis can exert important effects in distant target tissues via specific microbes and metabolites. Multiple studies have examined how diet-induced obese state is associated with gut dysbiosis and how gut microbes direct various physiological processes that help maintain obese state in a bidirectional crosstalk. Another tightly linked factor is sustained low grade inflammation in tumor microenvironment that is modulated by both obese state and dysbiosis, and influences tumor growth as well as response to immunotherapy. Our review brings together these important aspects and explores their connections. In this review, we discuss how obese state modulates various components of the breast tumor microenvironment and gut microbiota to achieve sustained low-grade inflammation. We explore the crosstalk between different components of tumor microenvironment and microbes, and how they might modulate the response to immunotherapy. Discussing studies from multiple tumor types, we delve to find common microbial characteristics that may positively or negatively influence immunotherapy efficacy in breast cancer and may guide future studies.


Sujet(s)
Dysbiose , Microbiome gastro-intestinal , Immunothérapie , Inflammation , Obésité , Microenvironnement tumoral , Humains , Dysbiose/immunologie , Obésité/immunologie , Obésité/thérapie , Obésité/microbiologie , Microenvironnement tumoral/immunologie , Microbiome gastro-intestinal/immunologie , Inflammation/immunologie , Immunothérapie/méthodes , Animaux , Tumeurs du sein/immunologie , Tumeurs du sein/thérapie , Femelle
14.
Front Immunol ; 15: 1412781, 2024.
Article de Anglais | MEDLINE | ID: mdl-39253092

RÉSUMÉ

Background: Autophagy plays important roles in cancer progression and therapeutic resistance, and the autophagy underlying the tumor pathogenesis and further mechanisms of chemoresistance emergence remains unknown. Methods: In this study, via the single-sample gene set enrichment analysis (ssGSEA) method, an autophagy 45-gene list was identified to evaluate samples' autophagy activity, verified through six GEO datasets with a confirmed autophagy phenotype. It was further utilized to distinguish tumors into autophagy score-high and score-low subtypes, and analyze their transcriptome landscapes, including survival analysis, correlation analysis of autophagy- and resistance-related genes, biological functional enrichment, and immune- and hypoxia-related and genomic heterogeneity comparison, in TCGA pan-cancer datasets. Furthermore, we performed an analysis of autophagy status in breast cancer chemoresistance combined with multiple GEO datasets and in vitro experiments to validate the mechanisms of potential anticancer drugs for reversing chemoresistance, including CCK-8 cell viability assays, RT-qPCR, and immunofluorescence. Results: The 45-gene list was used to identify autophagy score-high and score-low subtypes and further analyze their multi-dimensional features. We demonstrated that cancer autophagy status correlated with significantly different prognoses, molecular alterations, biological process activations, immunocyte infiltrations, hypoxia statuses, and specific mutational processes. The autophagy score-low subtype displayed a more favorable prognosis compared with the score-high subtype, associated with their immune-activated features, manifested as high immunocyte infiltration, including high CD8+T, Tfh, Treg, NK cells, and tumor-associated macrophages M1/M2. The autophagy score-low subtype also showed a high hypoxia score, and hypoxic tumors showed a significantly differential prognosis in different autophagy statuses. Therefore, "double-edged" cell fates triggered by autophagy might be closely correlated with the immune microenvironment and hypoxia induction. Results demonstrated that dysregulated autophagy was involved in many cancers and their therapeutic resistance and that the autophagy was induced by the resistance-reversing drug response, in five breast cancer GEO datasets and validated by in vitro experiments. In vitro, dihydroartemisinin and artesunate could reverse breast cancer doxorubicin resistance, through inducing autophagy via upregulating LC3B and ATG7. Conclusion: Our study provided a comprehensive landscape of the autophagy-related molecular and tumor microenvironment patterns for cancer progression and resistance, and highlighted the promising potential of drug-induced autophagy in the activation of drug sensitivity and reversal of resistance.


Sujet(s)
Autophagie , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Humains , Autophagie/génétique , Autophagie/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Lignée cellulaire tumorale , Microenvironnement tumoral/immunologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/immunologie , Tumeurs/anatomopathologie , Analyse de profil d'expression de gènes , Transcriptome , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Pronostic , Marqueurs biologiques tumoraux/génétique , Bases de données génétiques
15.
Cancer Biol Ther ; 25(1): 2398285, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39238191

RÉSUMÉ

Breast cancer stands as the most prevalent cancer diagnosed worldwide, often leading to brain metastasis, a challenging complication characterized by high mortality rates and a grim prognosis. Understanding the intricate mechanisms governing breast cancer brain metastasis (BCBM) remains an ongoing challenge. The unique microenvironment in the brain fosters an ideal setting for the colonization of breast cancer cells. The tumor microenvironment (TME) in brain metastases plays a pivotal role in the initiation and progression of BCBM, shaping the landscape for targeted therapeutic interventions. Current research primarily concentrates on unraveling the complexities of the TME in BCBM, with a particular emphasis on neuroglia and immune cells, such as microglia, monocyte-derived macrophages (MDMs), astrocytes and T cells. This comprehensive review delves deeply into these elements within the TME of BCBM, shedding light on their interplay, mechanisms, and potential as therapeutic targets to combat BCBM.


Sujet(s)
Tumeurs du cerveau , Tumeurs du sein , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Tumeurs du cerveau/secondaire , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/anatomopathologie , Femelle , Névroglie/anatomopathologie , Névroglie/immunologie , Animaux
16.
Front Immunol ; 15: 1396777, 2024.
Article de Anglais | MEDLINE | ID: mdl-39224600

RÉSUMÉ

Inflammation plays a pivotal role in cancer development, with chronic inflammation promoting tumor progression and treatment resistance, whereas acute inflammatory responses contribute to protective anti-tumor immunity. Gasdermin D (GSDMD) mediates the release of pro-inflammatory cytokines such as IL-1ß. While the release of IL-1ß is directly linked to the progression of several types of cancers, the role of GSDMD in cancer is less clear. In this study, we show that GSDMD expression is upregulated in human breast, kidney, liver, and prostate cancer. Higher GSDMD expression correlated with increased survival in primary breast invasive carcinoma (BRCA), but not in liver hepatocellular carcinoma (LIHC). In BRCA, but not in LIHC, high GSDMD expression correlated with a myeloid cell signature associated with improved prognosis. To further investigate the role of GSDMD in anticancer immunity, we induced breast cancer and hepatoma tumors in GSDMD-deficient mice. Contrary to our expectations, GSDMD deficiency had no effect on tumor growth, immune cell infiltration, or cytokine expression in the tumor microenvironment, except for Cxcl10 upregulation in hepatoma tumors. In vitro and in vivo innate immune activation with TLR ligands, that prime inflammatory responses, revealed no significant difference between GSDMD-deficient and wild-type mice. These results suggest that the impact of GSDMD on anticancer immunity is dependent on the tumor type. They underscore the complex role of inflammatory pathways in cancer, emphasizing the need for further exploration into the multifaceted effects of GSDMD in various tumor microenvironments. As several pharmacological modulators of GSDMD are available, this may lead to novel strategies for combination therapy in cancer.


Sujet(s)
Tumeurs du sein , Protéines et peptides de signalisation intracellulaire , Protéines de liaison aux phosphates , Microenvironnement tumoral , Animaux , Protéines de liaison aux phosphates/métabolisme , Protéines de liaison aux phosphates/génétique , Femelle , Humains , Souris , Tumeurs du sein/immunologie , Tumeurs du sein/mortalité , Tumeurs du sein/génétique , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Microenvironnement tumoral/immunologie , Souris knockout , Modèles animaux de maladie humaine , Lignée cellulaire tumorale , Cytokines/métabolisme , Tumeurs du foie/immunologie , Tumeurs du foie/mortalité , Tumeurs du foie/génétique , Gasdermines
17.
Breast Cancer Res ; 26(1): 129, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39232806

RÉSUMÉ

BACKGROUND: The internal heterogeneity of breast cancer, notably the tumor microenvironment (TME) consisting of malignant and non-malignant cells, has been extensively explored in recent years. The cells in this complex cellular ecosystem activate or suppress tumor immunity through phenotypic changes, secretion of metabolites and cell-cell communication networks. Macrophages, as the most abundant immune cells within the TME, are recruited by malignant cells and undergo phenotypic remodeling. Tumor-associated macrophages (TAMs) exhibit a variety of subtypes and functions, playing significant roles in impacting tumor immunity. However, their precise subtype delineation and specific function remain inadequately defined. METHODS: The publicly available single-cell transcriptomes of 49,141 cells from eight breast cancer patients with different molecular subtypes and stages were incorporated into our study. Unsupervised clustering and manual cell annotation were employed to accurately classify TAM subtypes. We then conducted functional analysis and constructed a developmental trajectory for TAM subtypes. Subsequently, the roles of TAM subtypes in cell-cell communication networks within the TME were explored using endothelial cells (ECs) and T cells as key nodes. Finally, analyses were repeated in another independent publish scRNA datasets to validate our findings for TAM characterization. RESULTS: TAMs are accurately classified into 7 subtypes, displaying anti-tumor or pro-tumor roles. For the first time, we identified a new TAM subtype capable of proliferation and expansion in breast cancer-TUBA1B+ TAMs playing a crucial role in TAMs diversity and tumor progression. The developmental trajectory illustrates how TAMs are remodeled within the TME and undergo phenotypic and functional changes, with TUBA1B+ TAMs at the initial point. Notably, the predominant TAM subtypes varied across different molecular subtypes and stages of breast cancer. Additionally, our research on cell-cell communication networks shows that TAMs exert effects by directly modulating intrinsic immunity, indirectly regulating adaptive immunity through T cells, as well as influencing tumor angiogenesis and lymphangiogenesis through ECs. CONCLUSIONS: Our study establishes a precise single-cell atlas of breast cancer TAMs, shedding light on their multifaceted roles in tumor biology and providing resources for targeting TAMs in breast cancer immunotherapy.


Sujet(s)
Tumeurs du sein , Analyse sur cellule unique , Transcriptome , Microenvironnement tumoral , Macrophages associés aux tumeurs , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Femelle , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Communication cellulaire/immunologie , Marqueurs biologiques tumoraux/génétique , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie
18.
Cancer Immunol Immunother ; 73(11): 223, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39235656

RÉSUMÉ

BACKGROUND: To assess the distribution characteristics of immune infiltration and lymphovascular invasion in breast cancer skin recurrence patients. METHODS: We retrospectively analyzed the clinicopathological data of patients who underwent radical surgery for primary breast cancer and experienced skin recurrence between January 2001 and April 2019. Immune and lymphovascular biomarkers were quantified in primary breast cancers, skin lesions and visceral metastatic lesions. Differences in biomarkers distribution between matched tissues were statistically analyzed using the Wilcoxon signed-rank test and Kruskal-Wallis one-way ANOVA. RESULTS: A total of 71 female breast cancer patients were reviewed in this study. Our study found that the expression levels of various lymphocyte immune markers in primary tumor specimens were higher than those in skin recurrences. The expression of CD8, CD57 and CD31 in primary breast cancer was higher than those in the skin. Compared to visceral metastatic lesions, D2-40 was highly expressed in the skin, while CD8 tended to decrease. In the skin specimens, the expression of CD8 (P < 0.001), FOXP3 (P = 0.006) and CD68 (P < 0.001) in the intratumoral area was higher, while the expression of CD57 (P < 0.001) was higher in the peritumoral area. Analyzing specimens from the same patient at different time points of skin progression, it was found that the expression of peritumoral CD4 decreased (P = 0.044) as the disease progressed. The low expression of D2-40 and CD163 in the skin lesions suggested a decrease in DFS. CONCLUSION: The immune microenvironment of breast cancer skin recurrence may be in a state of suppression, and this suppression may intensify with disease progression. The pattern of skin recurrence may be more inclined toward lymphatic invasion. Our study provides new insights into the biological behaviors of this disease and its response to immunotherapy.


Sujet(s)
Tumeurs du sein , Lymphocytes TIL , Récidive tumorale locale , Tumeurs cutanées , Humains , Femelle , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Tumeurs du sein/métabolisme , Tumeurs cutanées/immunologie , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/métabolisme , Adulte d'âge moyen , Études rétrospectives , Récidive tumorale locale/immunologie , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/métabolisme , Sujet âgé , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Adulte , Métastase lymphatique/anatomopathologie , Métastase lymphatique/immunologie , Marqueurs biologiques tumoraux/métabolisme , Microenvironnement tumoral/immunologie , Invasion tumorale , Pronostic
19.
J Immunother Cancer ; 12(8)2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39134345

RÉSUMÉ

BACKGROUND: It is encountering the dilemma of lacking precise biomarkers to predict the response to neoadjuvant chemoimmunotherapy (NACI) and determine whether patients should use immune checkpoint inhibitors (ICIs) in early breast cancer (BC). We aimed to develop a gene signature to predict NACI response for BC patients and identify individuals suitable for adding ICIs. PATIENTS AND METHODS: Two I-SPY2 cohorts and one West China Hospital cohort of patients treated with NACI were included. Machine learning algorithms were used to identify key genes. Principal component analysis was used to calculate the ImPredict (IP) score. The interaction effects between biomarkers and treatment regimens were examined based on the logistic regression analysis. The relationship between the IP score and immune microenvironment was investigated through immunohistochemistry (IHC) and multiplex IHC. RESULTS: The area under the curves of the IP score were 0.935, 0.865, and 0.841 in the discovery cohort, validation cohort 1, and in-house cohort. Marker-treatment interaction tests indicated that the benefits from immunotherapy significantly varied between patients with high and low IP scores (p for interaction <0.001), and patients with high IP scores were more suitable for immunotherapy addition. CONCLUSIONS: Our IP model shows favorable performance in predicting NACI response and is an effective tool for identifying BC patients who will benefit from ICIs. It may help clinicians optimize treatment strategies and guide clinical decision-making.


Sujet(s)
Tumeurs du sein , Immunothérapie , Traitement néoadjuvant , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/immunologie , Tumeurs du sein/génétique , Traitement néoadjuvant/méthodes , Adulte d'âge moyen , Immunothérapie/méthodes , Récepteur ErbB-2/métabolisme , Adulte , Marqueurs biologiques tumoraux , Sujet âgé
20.
Eur J Med Res ; 29(1): 414, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39135107

RÉSUMÉ

BACKGROUND: Breast cancer (BC), a common malignant tumor originating from the terminal ductal lobular unit of the breast, poses a substantial health risk to women. Previous studies have associated cytochrome b561 (CYB561) with a poor prognosis in BC; however, its underlying mechanism of this association remains unclear. METHODS: We investigated the expression of CYB561 mRNA in BC using databases such as The Cancer Genome Atlas, Gene Expression Omnibus, Tumor-Normal-Metastatic plot, and Kaplan-Meier plotter databases. The prognostic value of CYB561 protein in BC was assessed in relation to its expression levels in tumor tissue samples from 158 patients with BC. The effect of CYB561 on BC progression was confirmed using in vivo and in vitro experiments. The biological functions and related signaling pathways of CYB561 in BC were explored using gene microarray, Innovative Pathway, Gene Ontology enrichment, and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. The correlation between CYB561 and the BC tumor immune microenvironment was evaluated using the CIBERSORT algorithm and single-cell analysis and further validated through immunohistochemistry of serial sections. RESULTS: Our study demonstrated that upregulation of CYB561 expression predicted poor prognosis in patients with BC and that CYB561 knockdown inhibited the proliferation, migration, and invasive ability of BC cells in vitro. CYB561 knockdown inhibited BC tumor formation in vivo.CYB561 was observed to modulate downstream tropomyosin 1 expression. Furthermore, CYB561 expression was associated with macrophage M2 polarization in the BC immune microenvironment. CONCLUSIONS: Elevated CYB561 expression suggests a poor prognosis for patients with BC and is associated with macrophage M2 polarization in the BC microenvironment. Therefore, CYB561 could potentially serve as a therapeutic target for BC treatment.


Sujet(s)
Tumeurs du sein , Microenvironnement tumoral , Animaux , Femelle , Humains , Souris , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/immunologie , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Pronostic , Microenvironnement tumoral/immunologie , Protéines membranaires/génétique , Protéines membranaires/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE