Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 43
Filtrer
1.
Int Immunopharmacol ; 113(Pt A): 109420, 2022 Dec.
Article de Anglais | MEDLINE | ID: mdl-36461607

RÉSUMÉ

OBJECTIVE: Intercellular communication in the tumor microenvironment is a potential regulator of metastasis. To explore the specific mechanism, we performed a multi-omics analysis of hepatocellular carcinoma. MATERIALS AND METHODS: Multiple omics data including scRNA-seq, ATAC-seq, RNA-seq, and methylation data were obtained from GEO and TCGA databases. SCENIC was used to identify key transcription factors and their Regulatory networks. ScMLnet was used to explore the mechanism of intercellular communication in the microenvironment. Multiple omics studies based on RNA-seq, ATAC-seq, and methylation data were used to explore downstream mechanisms of key transcription factors. Based on the analysis of cell differentiation trajectory and transcription subtypes, the regulation of cell communication on tumor subtypes was studied, and possible therapeutic compounds were explored. The universality of this mechanism was investigated by post-Pan-cancer analysis. RESULTS: JUN and its regulatory network play a key role in HCC, which was mainly positively correlated with tumor-associated macrophages and fibroblasts. Intercellular communication analysis showed that macrophage and fibroblast-derived FN1 could increase JUN by TNFRSF11B/SMAD3. Multiomics analysis showed that KIF13A was a key downstream gene of JUN, which was involved in the activation of the hippo pathway. Analysis of cell differentiation trajectory, transcriptome subtypes, and neural network modeling showed that intercellular communication in the microenvironment can regulate the transcriptome characterization of HCC. Pan-cancer analysis indicates that this mechanism may be universal. CONCLUSION: FN1 derived from tumor-associated macrophages and fibroblasts promotes metastasis and alters transcriptome subtypes through the JUN-Hippo signaling pathway in HCC, which may be universal in cancers.


Sujet(s)
Carcinome hépatocellulaire , Fibroblastes , Tumeurs du foie , Macrophages associés aux tumeurs , Humains , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/physiopathologie , Fibroblastes/métabolisme , Fibroblastes/physiologie , Fibronectines/génétique , Fibronectines/métabolisme , Kinésine/génétique , Kinésine/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/physiopathologie , Microenvironnement tumoral/génétique , Microenvironnement tumoral/physiologie , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/physiologie
2.
Elife ; 112022 02 14.
Article de Anglais | MEDLINE | ID: mdl-35156921

RÉSUMÉ

The pancreatic ductal adenocarcinoma microenvironment is composed of a variety of cell types and marked by extensive fibrosis and inflammation. Tumor-associated macrophages (TAMs) are abundant, and they are important mediators of disease progression and invasion. TAMs are polarized in situ to a tumor promoting and immunosuppressive phenotype via cytokine signaling and metabolic crosstalk from malignant epithelial cells and other components of the tumor microenvironment. However, the specific distinguishing features and functions of TAMs remain poorly defined. Here, we generated tumor-educated macrophages (TEMs) in vitro and performed detailed, multiomic characterization (i.e., transcriptomics, proteomics, metabolomics). Our results reveal unique genetic and metabolic signatures of TEMs, the veracity of which were queried against our in-house single-cell RNA sequencing dataset of human pancreatic tumors. This analysis identified expression of novel, metabolic TEM markers in human pancreatic TAMs, including ARG1, ACLY, and TXNIP. We then utilized our TEM model system to study the role of mutant Kras signaling in cancer cells on TEM polarization. This revealed an important role for granulocyte-macrophage colony-stimulating factor (GM-CSF) and lactate on TEM polarization, molecules released from cancer cells in a mutant Kras-dependent manner. Lastly, we demonstrate that GM-CSF dysregulates TEM gene expression and metabolism through PI3K-AKT pathway signaling. Collectively, our results define new markers and programs to classify pancreatic TAMs, how these are engaged by cancer cells, and the precise signaling pathways mediating polarization.


Sujet(s)
Facteur de stimulation des colonies de granulocytes et de macrophages/métabolisme , Voies et réseaux métaboliques/immunologie , Tumeurs du pancréas/immunologie , Transduction du signal , Facteurs de transcription/métabolisme , Macrophages associés aux tumeurs/physiologie , Animaux , Lignée cellulaire tumorale , Transformation cellulaire néoplasique , Analyse de profil d'expression de gènes/méthodes , Humains , Métabolomique/méthodes , Souris , Souris de lignée C57BL , Tumeurs du pancréas/physiopathologie , Protéomique/méthodes , Macrophages associés aux tumeurs/immunologie
3.
Nat Commun ; 13(1): 626, 2022 02 02.
Article de Anglais | MEDLINE | ID: mdl-35110548

RÉSUMÉ

Metastases are initiated by disseminated tumor cells (DTCs) that colonize distant organs. Growing evidence suggests that the microenvironment of the primary tumor primes DTCs for dormant or proliferative fates. However, the manner in which this occurs remains poorly understood. Here, using the Window for High-Resolution Intravital Imaging of the Lung (WHRIL), we study the live lung longitudinally and follow the fate of individual DTCs that spontaneously disseminate from orthotopic breast tumors. We find that spontaneously DTCs have increased levels of retention, increased speed of extravasation, and greater survival after extravasation, compared to experimentally metastasized tumor cells. Detailed analysis reveals that a subset of macrophages within the primary tumor induces a pro-dissemination and pro-dormancy DTC phenotype. Our work provides insight into how specific primary tumor microenvironments prime a subpopulation of cells for expression of proteins associated with dissemination and dormancy.


Sujet(s)
Microenvironnement tumoral/physiologie , Macrophages associés aux tumeurs/physiologie , Animaux , Tumeurs du sein/génétique , Lignée cellulaire tumorale , Femelle , Humains , Souris , Souris de lignée C57BL , Tumeurs expérimentales , Cellules souches tumorales , Phénotype
4.
Theranostics ; 12(2): 963-975, 2022.
Article de Anglais | MEDLINE | ID: mdl-34976223

RÉSUMÉ

Rationale: Tumor-associated macrophages (TAMs), generally displaying the pro-tumor M2-like phenotype, strongly influence the progression of colorectal cancer (CRC) via their immunosuppressive activities. The high-mobility gene group A2 (HMGA2), an oncoprotein, is aberrantly overexpressed in CRC cells. However, the mechanisms by which tumor-derived HMGA2 modulates tumor microenvironment in CRC remain poorly understood. Methods:In vivo subcutaneous tumor xenograft model, azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced tumor mouse model and in vitro co-culture assays were used to investigate the Hmga2 role in TAM recruitment and polarization. Luciferase and chromatin immunoprecipitation (ChIP) assays were applied to examine the mechanism of HMGA2-mediated transcriptional regulation of signal transducer and activator of transcription 3 (STAT3). The CD68 correlation with patient outcome was analyzed in 167 human CRC tissues. Results: We found that HMGA2 in cancer cells promoted macrophage recruitment and M2 polarization in vitro and in vivo. HMGA2 directly bound to the STAT3 promoter to activate its transcription and subsequently induced CCL2 secretion, thus promoting macrophage recruitment. Our results from human CRC specimens also revealed a strong positive association between HMGA2 expression in tumor cells and CD68 expression in the stroma. We further showed that patients with an elevated CD68 expression had an unfavorable overall survival in all of the patients or in the subgroup with negative distant metastasis. Conclusion: Our work uncovers new insight into the link between the HMGA2/STAT3/CCL2 axis and macrophage recruitment in CRC. These findings provide a novel therapeutic option for targeting the HMGA2/STAT3/CCL2 axis in CRC.


Sujet(s)
Tumeurs colorectales/immunologie , Protéine HMGA2/métabolisme , Facteur de transcription STAT-3/métabolisme , Macrophages associés aux tumeurs/physiologie , Animaux , Polarité de la cellule , Chimiokine CCL2/métabolisme , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Techniques de knock-out de gènes , Cellules HEK293 , Humains , Muqueuse intestinale/métabolisme , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Régions promotrices (génétique) , Facteur de transcription STAT-3/génétique
5.
Int Immunopharmacol ; 103: 108451, 2022 Feb.
Article de Anglais | MEDLINE | ID: mdl-34923423

RÉSUMÉ

Glioblastoma (GBM, WHO grade 4) is a highly heterogeneous and aggressive primary malignant brain tumor. BTB domain and CNC homology 1 (BACH1) is a transcription factor, and it plays an essential role in regulating tumor metastasis, tumor metabolism, and tumor stem cell self-renewal. However, its role in glioma is still unclear. In this research, we confirmed that BACH1 as an independent prognostic indicator was enriched in GBMs. BACH1 was strongly correlated with immune responses in GBMs, especially the M0 and M2 tumor-associated macrophage (TAM) mediated immune responses. GBMs with high expression of BACH1 express high levels of immune checkpoints (ICs), glioma cell-derived TAM chemokines, and M2 TAM markers. Interestingly, single cell RNA-seq analysis showed that the expression level of BACH1 in TAMs was higher than that in the other cell types in GBM. Transcriptome analysis of U87-MG cells showed that compared with the BACH1-vector U87-MG group, glioma cell-derived TAM chemokines (including monocyte chemotactic protein-1 (MCP-1), granulocyte-macrophage colony-stimulating factor (GM-CSF), and EGF) and ICs (including CD276, TIM-3, LAG3, TIGIT and LGALS9) were enriched in the BACH1-overexpressing U87-MG group. In addition, we constructed a polygenic risk scoring model and compound nomogram model based on BACH1, which might provide a reliable prognosis assessment tool for clinicians and aid in treatment decision-making in the clinic. In conclusion, this research identified that BACH1 might be a potential molecular signature for survival and immunotherapy response. GBMs with high expression of BACH1 have a stronger immunosuppressive tumor microenvironment (TME). Overexpression of BACH1 can upregulate the expression of glioma cell-derived TAM chemokines and ICs in vitro. Moreover, the risk model and nomogram model based on BACH1 can provide a reliable prognosis assessment tool. Therefore, BACH1 is a promising therapeutic target for GBMs.


Sujet(s)
Facteurs de transcription à motif basique et à glissière à leucines/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du cerveau/métabolisme , Glioblastome/métabolisme , Immunothérapie/méthodes , Macrophages associés aux tumeurs/physiologie , Animaux , Facteurs de transcription à motif basique et à glissière à leucines/génétique , Marqueurs biologiques tumoraux/génétique , Tumeurs du cerveau/diagnostic , Tumeurs du cerveau/génétique , Lignée cellulaire tumorale , Chimiokines/métabolisme , Jeux de données comme sujet , Régulation de l'expression des gènes tumoraux , Glioblastome/diagnostic , Glioblastome/génétique , Humains , Activation des macrophages , Thérapie moléculaire ciblée , Nomogrammes , Risque , Analyse de survie
6.
Front Immunol ; 12: 778078, 2021.
Article de Anglais | MEDLINE | ID: mdl-34925351

RÉSUMÉ

Cellular associations in the bone microenvironment are involved in modulating the balance between bone remodeling and resorption, which is necessary for maintaining a normal bone morphology. Macrophages and osteoclasts are both vital components of the bone marrow. Macrophages can interact with osteoclasts and regulate bone metabolism by secreting a variety of cytokines, which make a significant contribution to the associations. Although, recent studies have fully explored either macrophages or osteoclasts, indicating the significance of these two types of cells. However, it is of high importance to report the latest discoveries on the relationships between these two myeloid-derived cells in the field of osteoimmunology. Therefore, this paper reviews this topic from three novel aspects of the origin, polarization, and subgroups based on the previous work, to provide a reference for future research and treatment of bone-related diseases.


Sujet(s)
Communication cellulaire , Macrophages/physiologie , Ostéoclastes/physiologie , Animaux , Polarité de la cellule , Cytokines/physiologie , Humains , Macrophages/classification , Monoxyde d'azote/physiologie , Ostéoclastes/classification , Espèces réactives de l'oxygène/métabolisme , Macrophages associés aux tumeurs/physiologie
7.
J Zhejiang Univ Sci B ; 22(11): 885-892, 2021 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-34783219

RÉSUMÉ

Osteosarcoma (OS) is the most common primary bone tumor in children and adolescents. It is an aggressive tumor with a tendency to spread to the lung, which is the most common site of metastasis. Patients with advanced OS with metastases have poor prognoses despite the application of chemotherapy, thus highlighting the need for novel therapeutic targets. The tumor microenvironment (TME) of OS is confirmed to be essential for and supportive of tumor growth and dissemination. The immune component of the OS microenvironment is mainly composed of tumor-associated macrophages (TAMs). In OS, TAMs promote tumor growth and angiogenesis and upregulate the cancer stem cell-like phenotype. However, TAMs inhibit the metastasis of OS. Therefore, much attention has been paid to investigating the mechanism of TAMs in OS development and the progression of immunotherapy for OS. In this article, we aim to summarize the roles of TAMs in OS and the major findings on the application of TAMs in OS treatment.


Sujet(s)
Tumeurs osseuses/immunologie , Ostéosarcome/immunologie , Macrophages associés aux tumeurs/physiologie , Tumeurs osseuses/vascularisation , Tumeurs osseuses/anatomopathologie , Humains , Métastase tumorale , Cellules souches tumorales/physiologie , Néovascularisation pathologique/étiologie , Ostéosarcome/vascularisation , Ostéosarcome/anatomopathologie , Microenvironnement tumoral/physiologie
8.
Bioengineered ; 12(1): 5462-5475, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34506209

RÉSUMÉ

Colorectal cancer (CRC) threatens the health of patients with high mortality, which lacks sensitive biomarkers for diagnosis to improve total survival. The lncRNA NBR2 is reported to be downregulated in CRC and suppresses the proliferation of CRC cells. However, the underlying mechanisms remain unclear. The present study aimed to explore the regulatory function of the lncRNA NBR2 on tumor-associated macrophage (TAM) polarization and its consequent anti-tumor effect. Two CRC cell lines were used in this study. We found that the lncRNA NBR2, TNF-α, and HLA-DR were downregulated, and Arg-1, CD163, CD206, and IL-4 were upregulated in CRC tumors. M1 polarization was activated and M2 polarization was suppressed in NBR2-overexpressed macrophages, accompanied by increased production of inflammatory factors, decreased proliferation, and inhibited migration ability in the co-culture system of HCT-116 cells (SW480 cells) and NBR2-overexpressed macrophages. The promoted proliferation and migration were observed in the co-culture system of HCT-116 cells (SW480 cells) and NBR2-knockdown macrophages. The tumor growth of both HCT-116 cells and SW480 cells in the xenograft model was suppressed by co-planting NBR2-overexpressed macrophages and was facilitated by the co-planting of NBR2-knockdown macrophages. The release of inflammatory factors was induced, M1 polarization was facilitated, and M2 polarization was suppressed in tumor tissues in the NBR2-overexpressed group, which were all reversed in the NBR2-knockdown group. Therefore, the lncRNA NBR2 suppressed the progression of colorectal cancer in vitro and in vivo by regulating TAM polarization.


Sujet(s)
Tumeurs colorectales , ARN long non codant , Facteurs de transcription , Macrophages associés aux tumeurs , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Évolution de la maladie , Femelle , Humains , Souris , Souris nude , ARN long non codant/génétique , ARN long non codant/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Macrophages associés aux tumeurs/cytologie , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/physiologie
9.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-34502382

RÉSUMÉ

Tumor-associated macrophages (TAMs) promote cancer cell proliferation and metastasis, as well as anti-tumor immune suppression. Recent studies have shown that tumors enhance the recruitment and differentiation of TAMs, but the detailed mechanisms have not been clarified. We thus examined the influence of cancer cells on the differentiation of monocytes to TAM subsets, including CD163+, CD204+, and CD206+ cells, in oral squamous cell carcinoma (OSCC) using immunohistochemistry, flow cytometry, and a cytokine array. Furthermore, we investigated the effect of OSCC cells (HSC-2, SQUU-A, and SQUU-B cells) on the differentiation of purified CD14+ cells to TAM subsets. The localization patterns of CD163+, CD204+, and CD206+ in OSCC sections were quite different. The expression of CD206 on CD14+ cells was significantly increased after the co-culture with OSCC cell lines, while the expressions of CD163 and CD204 on CD14+ cells showed no change. High concentrations of plasminogen activator inhibitor-1 (PAI-1) and interleukin-8 (IL-8) were detected in the conditioned medium of OSCC cell lines. PAI-1 and IL-8 stimulated CD14+ cells to express CD206. Moreover, there were positive correlations among the numbers of CD206+, PAI-1+, and IL-8+ cells in OSCC sections. These results suggest that PAI-1 and IL-8 produced by OSCC contribute to the differentiation of monocytes to CD206+ TAMs.


Sujet(s)
Macrophages/métabolisme , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde/anatomopathologie , Différenciation cellulaire/génétique , Lignée cellulaire tumorale , Tumeurs de la tête et du cou/anatomopathologie , Humains , Immunohistochimie , Interleukine-8/métabolisme , Interleukine-8/physiologie , Agranulocytes/cytologie , Macrophages/physiologie , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Inhibiteur-1 d'activateur du plasminogène/physiologie , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Microenvironnement tumoral , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/physiologie
10.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article de Anglais | MEDLINE | ID: mdl-34504016

RÉSUMÉ

Expression and function of odorant receptors (ORs), which account for more than 50% of G protein-coupled receptors, are being increasingly reported in nonolfactory sites. However, ORs that can be targeted by drugs to treat diseases remain poorly identified. Tumor-derived lactate plays a crucial role in multiple signaling pathways leading to generation of tumor-associated macrophages (TAMs). In this study, we hypothesized that the macrophage OR Olfr78 functions as a lactate sensor and shapes the macrophage-tumor axis. Using Olfr78+/+ and Olfr78-/- bone marrow-derived macrophages with or without exogenous Olfr78 expression, we demonstrated that Olfr78 sensed tumor-derived lactate, which was the main factor in tumor-conditioned media responsible for generation of protumoral M2-TAMs. Olfr78 functioned together with Gpr132 to mediate lactate-induced generation of protumoral M2-TAMs. In addition, syngeneic Olfr78-deficient mice exhibited reduced tumor progression and metastasis together with an increased anti- versus protumoral immune cell population. We propose that the Olfr78-lactate interaction is a therapeutic target to reduce and prevent tumor progression and metastasis.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Récepteurs olfactifs/métabolisme , Macrophages associés aux tumeurs/métabolisme , Animaux , Protéines du cycle cellulaire/physiologie , Lignée cellulaire tumorale , Femelle , Humains , Acide lactique/métabolisme , Macrophages/métabolisme , Mâle , Souris , Souris de lignée C57BL , Phénotype , Récepteurs couplés aux protéines G/physiologie , Récepteurs olfactifs/physiologie , Transduction du signal , Microenvironnement tumoral , Macrophages associés aux tumeurs/physiologie
11.
Cancer Sci ; 112(10): 4198-4207, 2021 Oct.
Article de Anglais | MEDLINE | ID: mdl-34375482

RÉSUMÉ

Tumor-associated macrophages (TAMs), one of the most common cell components in the tumor microenvironment, have been reported as key contributors to cancer-related inflammation and enhanced metastatic progression of tumors. To explore the underlying mechanism of TAM-induced tumor progression, TAMs were isolated from colorectal cancer patients, and the functional interaction with colorectal cancer cells was analyzed. Our study found that coculture of TAMs contributed to a glycolytic state in colorectal cancer, which promoted the stem-like phenotypes and invasion of tumor cells. TAMs produced the cytokine transforming growth factor-ß to support hypoxia-inducible factor 1α (HIF1α) expression, thereby upregulating Tribbles pseudokinase 3 (TRIB3) in tumor cells. Elevated expression of TRIB3 resulted in activation of the ß-catenin/Wnt signaling pathway, which eventually enhanced the stem-like phenotypes and cell invasion in colorectal cancer. Our findings provided evidence that TAMs promoted colorectal cancer progression in a HIF1α/TRIB3-dependent manner, and blockade of HIF1α signals efficiently improved the outcome of chemotherapy, describing an innovative approach for colorectal cancer treatment.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Tumeurs colorectales/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protéines de répression/métabolisme , Facteur de croissance transformant bêta/physiologie , Macrophages associés aux tumeurs/physiologie , Animaux , Prolifération cellulaire , Techniques de coculture , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Évolution de la maladie , Femelle , Cellules HCT116 , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/antagonistes et inhibiteurs , Tumeurs du poumon/secondaire , Souris , Souris de lignée BALB C , Souris de lignée NOD , Souris SCID , Invasion tumorale , Cellules souches tumorales , Phénotype , Protein-Serine-Threonine Kinases/métabolisme , Facteur de croissance transformant bêta/métabolisme , Microenvironnement tumoral , Macrophages associés aux tumeurs/métabolisme , Régulation positive , Voie de signalisation Wnt/physiologie
12.
Cancer Sci ; 112(10): 4151-4165, 2021 Oct.
Article de Anglais | MEDLINE | ID: mdl-34293235

RÉSUMÉ

Submucosal invasion and lymph node metastasis are important issues affecting treatment options for early colorectal cancer (CRC). In this study, we aimed to unravel the molecular mechanism underlying the invasiveness of early CRCs. We performed RNA-sequencing (RNA-seq) with poorly differentiated components (PORs) and their normal counterparts isolated from T1 CRC tissues and detected significant upregulation of serum amyloid A1 (SAA1) in PORs. Immunohistochemical analysis revealed that SAA1 was specifically expressed in PORs at the invasive front of T1b CRCs. Upregulation of SAA1 in CRC cells promoted cell migration and invasion. Coculture experiments using CRC cell lines and THP-1 cells suggested that interleukin 1ß (IL-1ß) produced by macrophages induces SAA1 expression in CRC cells. Induction of SAA1 and promotion of CRC cell migration and invasion by macrophages were inhibited by blocking IL-1ß. These findings were supported by immunohistochemical analysis of primary T1 CRCs showing accumulation of M1-like/M2-like macrophages at SAA1-positive invasive front regions. Moreover, SAA1 produced by CRC cells stimulated upregulation of matrix metalloproteinase-9 in macrophages. Our data suggest that tumor-associated macrophages at the invasive front of early CRCs promote cancer cell migration and invasion through induction of SAA1 and that SAA1 may be a predictive biomarker and a useful therapeutic target.


Sujet(s)
Tumeurs colorectales/anatomopathologie , Interleukine-1 bêta/métabolisme , Protéine amyloïde A sérique/métabolisme , Macrophages associés aux tumeurs/physiologie , Sujet âgé , Séquence nucléotidique , Mouvement cellulaire , Techniques de coculture , Tumeurs colorectales/métabolisme , Femelle , Humains , Interleukine-1 bêta/antagonistes et inhibiteurs , Mâle , Matrix metalloproteinase 9/métabolisme , Adulte d'âge moyen , Invasion tumorale , Protéines tumorales/métabolisme , Cellules THP-1 , Macrophages associés aux tumeurs/métabolisme , Régulation positive
13.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-34298975

RÉSUMÉ

Previously, we showed that chemotherapy paradoxically exacerbated cancer cell colonization at the secondary site in a manner dependent on Atf3, a stress-inducible gene, in the non-cancer host cells. Here, we present evidence that this phenotype is established at an early stage of colonization within days of cancer cell arrival. Using mouse breast cancer models, we showed that, in the wild-type (WT) lung, cyclophosphamide (CTX) increased the ability of the lung to retain cancer cells in the vascular bed. Although CTX did not change the WT lung to affect cancer cell extravasation or proliferation, it changed the lung macrophage to be pro-cancer, protecting cancer cells from death. This, combined with the initial increase in cell retention, resulted in higher lung colonization in CTX-treated than control-treated mice. In the Atf3 knockout (KO) lung, CTX also increased the ability of lung to retain cancer cells. However, the CTX-treated KO macrophage was highly cytotoxic to cancer cells, resulting in no increase in lung colonization-despite the initial increase in cell retention. In summary, the status of Atf3 dictates the dichotomous activity of macrophage: pro-cancer for CTX-treated WT macrophage but anti-cancer for the KO counterpart. This dichotomy provides a mechanistic explanation for CTX to exacerbate lung colonization in the WT but not Atf3 KO lung.


Sujet(s)
Facteur de transcription ATF-3/physiologie , Cyclophosphamide/toxicité , Tumeurs du poumon/secondaire , Macrophages/physiologie , Tumeurs expérimentales de la mamelle/génétique , Métastase tumorale/physiopathologie , Protéines tumorales/physiologie , Stress physiologique/génétique , Macrophages associés aux tumeurs/physiologie , Animaux , Peptides antimicrobiens cationiques/biosynthèse , Peptides antimicrobiens cationiques/génétique , Lignée cellulaire tumorale , Cyclophosphamide/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Gènes rapporteurs , Génotype , Humains , Tumeurs du poumon/métabolisme , Activation des macrophages , Tumeurs expérimentales de la mamelle/traitement médicamenteux , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris de lignée C57BL , Lignées consanguines de souris , Souris knockout , Souris transgéniques , Traitement néoadjuvant/effets indésirables , Métastase tumorale/génétique , Protéines tumorales/biosynthèse , Protéines tumorales/génétique , Transplantation tumorale/méthodes , Cellules souches tumorales/anatomopathologie , Migration transendothéliale et transépithéliale , Microenvironnement tumoral , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Cathélicidines
14.
Cell ; 184(11): 2988-3005.e16, 2021 05 27.
Article de Anglais | MEDLINE | ID: mdl-34019793

RÉSUMÉ

Clear cell renal carcinoma (ccRCC) is a heterogeneous disease with a variable post-surgical course. To assemble a comprehensive ccRCC tumor microenvironment (TME) atlas, we performed single-cell RNA sequencing (scRNA-seq) of hematopoietic and non-hematopoietic subpopulations from tumor and tumor-adjacent tissue of treatment-naive ccRCC resections. We leveraged the VIPER algorithm to quantitate single-cell protein activity and validated this approach by comparison to flow cytometry. The analysis identified key TME subpopulations, as well as their master regulators and candidate cell-cell interactions, revealing clinically relevant populations, undetectable by gene-expression analysis. Specifically, we uncovered a tumor-specific macrophage subpopulation characterized by upregulation of TREM2/APOE/C1Q, validated by spatially resolved, quantitative multispectral immunofluorescence. In a large clinical validation cohort, these markers were significantly enriched in tumors from patients who recurred following surgery. The study thus identifies TREM2/APOE/C1Q-positive macrophage infiltration as a potential prognostic biomarker for ccRCC recurrence, as well as a candidate therapeutic target.


Sujet(s)
Néphrocarcinome/métabolisme , Récidive tumorale locale/génétique , Macrophages associés aux tumeurs/métabolisme , Adulte , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Marqueurs biologiques tumoraux/génétique , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Études de cohortes , Femelle , Expression des gènes/génétique , Régulation de l'expression des gènes tumoraux/génétique , Humains , Rein/métabolisme , Tumeurs du rein/anatomopathologie , Lymphocytes TIL/anatomopathologie , Macrophages/métabolisme , Mâle , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Adulte d'âge moyen , Récidive tumorale locale/métabolisme , Pronostic , Récepteurs au complément/génétique , Récepteurs au complément/métabolisme , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes , Microenvironnement tumoral , Macrophages associés aux tumeurs/physiologie
15.
Cancer Sci ; 112(7): 2692-2704, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-33964093

RÉSUMÉ

Macrophages are an essential component of antitumor activity; however, the role of tumor-associated macrophages (TAMs) in colorectal cancer (CRC) remains controversial. Here, we elucidated the role of TAMs in CRC progression, especially at the early stage. We assessed the TAM number, phenotype, and distribution in 53 patients with colorectal neoplasia, including intramucosal neoplasia, submucosal invasive colorectal cancer (SM-CRC), and advanced cancer, using double immunofluorescence for CD68 and CD163. Next, we focused on the invasive front in SM-CRC and association between TAMs and clinicopathological features including lymph node metastasis, which were evaluated in 87 SM-CRC clinical specimens. The number of M2 macrophages increased with tumor progression and dynamic changes were observed with respect to the number and phenotype of TAMs at the invasive front, especially at the stage of submucosal invasion. A high M2 macrophage count at the invasive front was correlated with lymphovascular invasion, low histological differentiation, and lymph node metastasis; a low M1 macrophage count at the invasive front was correlated with lymph node metastasis. Furthermore, receiver operating characteristic curve analysis revealed that the M2/M1 ratio was a better predictor of the risk of lymph node metastasis than the pan-, M1, or M2 macrophage counts at the invasive front. These results suggested that TAMs at the invasive front might play a role in CRC progression, especially at the early stages. Therefore, evaluating the TAM phenotype, number, and distribution may be a potential predictor of metastasis, including lymph node metastasis, and TAMs may be a potential CRC therapeutic target.


Sujet(s)
Tumeurs colorectales/anatomopathologie , Macrophages associés aux tumeurs/physiologie , Sujet âgé , Antigènes CD/analyse , Antigènes de différenciation des myélomonocytes/analyse , Numération cellulaire , Différenciation cellulaire , Tumeurs colorectales/immunologie , Évolution de la maladie , Transition épithélio-mésenchymateuse , Femelle , Technique d'immunofluorescence/méthodes , Humains , Métastase lymphatique , Mâle , Invasion tumorale , Phénotype , Courbe ROC , Récepteurs de surface cellulaire/analyse , Microenvironnement tumoral , Macrophages associés aux tumeurs/cytologie
16.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article de Anglais | MEDLINE | ID: mdl-33846242

RÉSUMÉ

Precision medicine in oncology leverages clinical observations of exceptional response. Toward an understanding of the molecular features that define this response, we applied an integrated, multiplatform analysis of RNA profiles derived from clinically annotated glioblastoma samples. This analysis suggested that specimens from exceptional responders are characterized by decreased accumulation of microglia/macrophages in the glioblastoma microenvironment. Glioblastoma-associated microglia/macrophages secreted interleukin 11 (IL11) to activate STAT3-MYC signaling in glioblastoma cells. This signaling induced stem cell states that confer enhanced tumorigenicity and resistance to the standard-of-care chemotherapy, temozolomide (TMZ). Targeting a myeloid cell restricted an isoform of phosphoinositide-3-kinase, phosphoinositide-3-kinase gamma isoform (PI3Kγ), by pharmacologic inhibition or genetic inactivation disrupted this signaling axis by reducing microglia/macrophage-associated IL11 secretion in the tumor microenvironment. Mirroring the clinical outcomes of exceptional responders, PI3Kγ inhibition synergistically enhanced the anti-neoplastic effects of TMZ in orthotopic murine glioblastoma models. Moreover, inhibition or genetic inactivation of PI3Kγ in murine glioblastoma models recapitulated expression profiles observed in clinical specimens isolated from exceptional responders. Our results suggest key contributions from tumor-associated microglia/macrophages in exceptional responses and highlight the translational potential for PI3Kγ inhibition as a glioblastoma therapy.


Sujet(s)
Glioblastome/métabolisme , Microglie/métabolisme , Témozolomide/pharmacologie , Adulte , Animaux , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Résistance aux médicaments antinéoplasiques/physiologie , Femelle , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Humains , Interleukine-11/immunologie , Interleukine-11/métabolisme , Mâle , Souris de lignée C57BL , Souris nude , Microglie/physiologie , Phosphatidylinositol 3-kinase/métabolisme , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Témozolomide/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/physiologie
17.
Biosci Rep ; 41(5)2021 05 28.
Article de Anglais | MEDLINE | ID: mdl-33928349

RÉSUMÉ

Tumor microenvironment (TME) plays a particularly important role in the progression, invasion and metastasis of cervical carcinoma (CC). Tumor-associated macrophages (TAMs) are significant components of the tumor microenvironment in CC. However, the results of studies on the correlation between TAMs and progression in CC are still controversial. This research aimed to investigate the relationship between TAMs infiltration and progression in CC. A total of 100 patients with CC were included in the study. The correlation between TAMs and clinicopathologic features was studied. Besides, a systematic literature search was conducted from legitimate electronic databases to specifically evaluate the role of TAMs in TME of cervical carcinoma. In the meta-analysis, high stromal CD68+ TAMs density was relevant to lymph node metastasis (WMD = 11.89, 95% CI: 5.30-18.47). At the same time, CD163+ M2 TAM density was associated with lymph node metastasis (OR = 2.42, 95% CI: 1.09-5.37; WMD = 39.37, 95% CI: 28.25-50.49) and FIGO stage (WMD = -33.60, 95% CI: -45.04 to -22.16). This was further confirmed in the experimental study of 100 tissues of cervical cancer. It supported a critical role of TAMs as a prospective predictor of cervical cancer. In conclusion, CD68+ TAM and CD163+ M2 TAM infiltration in CC were associated with tumor progression. And CD163+ M2 TAM infiltration was associated with more advanced FIGO stage and lymph node metastasis in CC.


Sujet(s)
Carcinomes/anatomopathologie , Macrophages associés aux tumeurs/anatomopathologie , Tumeurs du col de l'utérus/anatomopathologie , Adulte , Antigènes CD/génétique , Antigènes CD/métabolisme , Mouvement cellulaire , Femelle , Humains , Métastase lymphatique/anatomopathologie , Adulte d'âge moyen , Microenvironnement tumoral , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/physiologie
18.
Theranostics ; 11(8): 3839-3852, 2021.
Article de Anglais | MEDLINE | ID: mdl-33664865

RÉSUMÉ

Background: Lactate greatly contributes to the regulation of intracellular communication within the tumor microenvironment (TME). However, the role of lactate in pituitary adenoma (PA) invasion is unclear. In this study, we aimed to clarify the effects of lactate on the TME and the effects of TME on PA invasion. Methods: To explore the correlation between TME acidosis and tumor invasion, LDHA and LAMP2 expression levels were quantified in invasive (n = 32) and noninvasive (n = 32) PA samples. The correlation between immune cell infiltration and tumor invasion was evaluated in 64 PAs. Critical chemokine and key signaling pathway components were detected by qPCR, Western blotting, siRNA knockdown, and specific inhibitors. The functional consequences of CCR4 signaling inhibition were evaluated in vitro and in vivo. Results: Lactate was positively associated with PA invasion. Of the 64 PA tissues, invasive PAs were related to high infiltration of M2-like tumor-associated macrophages (TAMs) (P < 0.05). Moreover, lactate secreted from PA cells facilitated M2 polarization via the mTORC2 and ERK signaling pathways, while activated TAMs secreted CCL17 to promote PA invasion via the CCL17/CCR4/mTORC1 axis. According to univariate analysis of clinical data, high CCL17 expression was associated with larger tumor size (P = 0.0438), greater invasion (P = 0.0334), and higher susceptibility to postoperative recurrence (P = 0.0195) in human PAs. Conclusion: This study illustrates the dynamics between PA cells and immune TME in promoting PA invasion via M2 polarization. CCL17 levels in the TME are related to the PA invasiveness and clinical prognosis, and the CCL17/CCR4/mTOCR1 axis may serve as potential therapeutic targets for Pas.


Sujet(s)
Adénomes/anatomopathologie , Adénomes/physiopathologie , Chimiokine CCL17/métabolisme , Acide lactique/métabolisme , Tumeurs de l'hypophyse/anatomopathologie , Tumeurs de l'hypophyse/physiopathologie , Macrophages associés aux tumeurs/physiologie , Adulte , Femelle , Humains , Acide lactique/pharmacologie , Mâle , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Adulte d'âge moyen , Modèles biologiques , Invasion tumorale , Récidive tumorale locale/métabolisme , Médecine de précision , Récepteurs CCR4/métabolisme , Transduction du signal , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/physiologie , Macrophages associés aux tumeurs/classification , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques
19.
Life Sci Alliance ; 4(6)2021 06.
Article de Anglais | MEDLINE | ID: mdl-33782087

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDA) is accompanied by reprogramming of the local microenvironment, but changes at distal sites are poorly understood. We implanted biomaterial scaffolds, which act as an artificial premetastatic niche, into immunocompetent tumor-bearing and control mice, and identified a unique tumor-specific gene expression signature that includes high expression of C1qa, C1qb, Trem2, and Chil3 Single-cell RNA sequencing mapped these genes to two distinct macrophage populations in the scaffolds, one marked by elevated C1qa, C1qb, and Trem2, the other with high Chil3, Ly6c2 and Plac8 In mice, expression of these genes in the corresponding populations was elevated in tumor-associated macrophages compared with macrophages in the normal pancreas. We then analyzed single-cell RNA sequencing from patient samples, and determined expression of C1QA, C1QB, and TREM2 is elevated in human macrophages in primary tumors and liver metastases. Single-cell sequencing analysis of patient blood revealed a substantial enrichment of the same gene signature in monocytes. Taken together, our study identifies two distinct tumor-associated macrophage and monocyte populations that reflects systemic immune changes in pancreatic ductal adenocarcinoma patients.


Sujet(s)
Monocytes/métabolisme , Tumeurs du pancréas/métabolisme , Macrophages associés aux tumeurs/métabolisme , Adulte , Animaux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Protéines de transport , Complément C1q , Femelle , Expression des gènes/génétique , Régulation de l'expression des gènes tumoraux/génétique , Humains , Macrophages/métabolisme , Mâle , Glycoprotéines membranaires/métabolisme , Souris , Souris de lignée C57BL , Protéines mitochondriales , Tumeurs du pancréas/sang , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Récepteurs au complément , Récepteurs immunologiques/métabolisme , Analyse de séquence d'ARN , Analyse sur cellule unique , Transcriptome/génétique , Microenvironnement tumoral/génétique , Macrophages associés aux tumeurs/physiologie , Tumeurs du pancréas
20.
Cancer Lett ; 506: 83-94, 2021 05 28.
Article de Anglais | MEDLINE | ID: mdl-33676940

RÉSUMÉ

Glioma stem cells (GSCs) are thought to underlie glioma initiation, evolution, resistance to therapies, and relapse. They are defined by their capacity to initiate glioma in immunocompromised mice which precludes analysis of their interaction with immune cells. Macrophages dominate the immune cell composition in glioma. We hypothesized that stemness and immune evasion induced by macrophages are closed intertwined in glioma. By using mass cytometry and RNA sequencing, we reveal that in immunocompetent mice, FGL2 promotes the stem-like phenotypes of glioma cells in an expression level-dependent manner. Mechanistically, FGL2-producing glioma cells recruit macrophages into the tumor microenvironment and induce the macrophages to secrete CXCL7 via the CD16/SyK/PI3K/HIF1α pathways. CXCL7, in turn, enhances the stem-like functionality of glioma cells, resulting in an increase in tumor incidence and progression that can be blocked with a neutralizing anti-CXCL7 antibody. Clinically, the FGL2-CXCL7 paracrine loop positively correlated with a higher macrophage signature and poorer prognosis in glioma patients. Thus, glioma cells' stem-like functionality is regulated by FGL2 in the presence of macrophages, and the FGL2-CXCL7 paracrine signaling axis is critical for regulating this function.


Sujet(s)
Tumeurs du cerveau/étiologie , Chimiokines CXC/physiologie , Fibrinogène/physiologie , Gliome/étiologie , Cellules souches tumorales/physiologie , Macrophages associés aux tumeurs/physiologie , Animaux , Tumeurs du cerveau/anatomopathologie , Carcinogenèse , Lignée cellulaire tumorale , Mouvement cellulaire , Gliome/anatomopathologie , Souris , Souris de lignée C57BL , Cellules myéloïdes/physiologie , Transduction du signal/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE