34 results on '"Yvon Elkrim"'
Search Results
2. Data from IL1β Promotes Immune Suppression in the Tumor Microenvironment Independent of the Inflammasome and Gasdermin D
- Author
-
Damya Laoui, Jo A. Van Ginderachter, Mohamed Lamkanfi, Andy Wullaert, Alain Beschin, Massimiliano Mazzone, Diether Lambrechts, Geert van Loo, Geert Raes, Louis Boon, Amelie Fossoul, Ayla Debraekeleer, Jan Brughmans, Maryse Schmoetten, Yvon Elkrim, Sana M. Arnouk, Pauline M.R. Bardet, Jiri Keirsse, Evangelia Bolli, Samantha Pretto, Manuel Ehling, Junbin Qian, Aleksandar Murgaski, Helena Van Damme, Els Lebegge, Pedro H.V. Saavedra, Lieselotte Vande Walle, and Máté Kiss
- Abstract
IL1β is a central mediator of inflammation. Secretion of IL1β typically requires proteolytic maturation by the inflammasome and formation of membrane pores by gasdermin D (GSDMD). Emerging evidence suggests an important role for IL1β in promoting cancer progression in patients, but the underlying mechanisms are ill-defined. Here, we have shown a key role for IL1β in driving tumor progression in two distinct mouse tumor models. Notably, activation of the inflammasome, caspase-8, as well as the pore-forming proteins GSDMD and mixed lineage kinase domain–like protein in the host were dispensable for the release of intratumoral bioactive IL1β. Inflammasome-independent IL1β release promoted systemic neutrophil expansion and fostered accumulation of T-cell–suppressive neutrophils in the tumor. Moreover, IL1β was essential for neutrophil infiltration triggered by antiangiogenic therapy, thereby contributing to treatment-induced immunosuppression. Deletion of IL1β allowed intratumoral accumulation of CD8+ effector T cells that subsequently activated tumor-associated macrophages. Depletion of either CD8+ T cells or macrophages abolished tumor growth inhibition in IL1β-deficient mice, demonstrating a crucial role for CD8+ T-cell–macrophage cross-talk in the antitumor immune response. Overall, these results support a tumor-promoting role for IL1β through establishing an immunosuppressive microenvironment and show that inflammasome activation is not essential for release of this cytokine in tumors.
- Published
- 2023
3. Data from Efficacy of CD40 Agonists Is Mediated by Distinct cDC Subsets and Subverted by Suppressive Macrophages
- Author
-
Damya Laoui, Carola H. Ries, Martina Schmittnaegel, Greetje Vande Velde, Abhishek D. Garg, Sophie Janssens, Sofie Deschoemaeker, Niels Vandamme, Sabine Hoves, Louis Boon, Yvon Elkrim, Victor Bosteels, Ayla Debraekeleer, Ahmed E.I. Hamouda, Sana M. Arnouk, Jan Brughmans, Eva Hadadi, Jiri Keirsse, Isaure Vanmeerbeek, Daliya Kancheva, Helena Van Damme, Máté Kiss, and Aleksandar Murgaski
- Abstract
Agonistic αCD40 therapy has been shown to inhibit cancer progression in only a fraction of patients. Understanding the cancer cell–intrinsic and microenvironmental determinants of αCD40 therapy response is therefore crucial to identify responsive patient populations and to design efficient combinatorial treatments. Here, we show that the therapeutic efficacy of αCD40 in subcutaneous melanoma relies on preexisting, type 1 classical dendritic cell (cDC1)–primed CD8+ T cells. However, after administration of αCD40, cDC1s were dispensable for antitumor efficacy. Instead, the abundance of activated cDCs, potentially derived from cDC2 cells, increased and further activated antitumor CD8+ T cells. Hence, distinct cDC subsets contributed to the induction of αCD40 responses. In contrast, lung carcinomas, characterized by a high abundance of macrophages, were resistant to αCD40 therapy. Combining αCD40 therapy with macrophage depletion led to tumor growth inhibition only in the presence of strong neoantigens. Accordingly, treatment with immunogenic cell death–inducing chemotherapy sensitized lung tumors to αCD40 therapy in subcutaneous and orthotopic settings. These insights into the microenvironmental regulators of response to αCD40 suggest that different tumor types would benefit from different combinations of therapies to optimize the clinical application of CD40 agonists.Significance:This work highlights the temporal roles of different dendritic cell subsets in promoting CD8+ T-cell–driven responses to CD40 agonist therapy in cancer.
- Published
- 2023
4. Supplementary Figure from Efficacy of CD40 Agonists Is Mediated by Distinct cDC Subsets and Subverted by Suppressive Macrophages
- Author
-
Damya Laoui, Carola H. Ries, Martina Schmittnaegel, Greetje Vande Velde, Abhishek D. Garg, Sophie Janssens, Sofie Deschoemaeker, Niels Vandamme, Sabine Hoves, Louis Boon, Yvon Elkrim, Victor Bosteels, Ayla Debraekeleer, Ahmed E.I. Hamouda, Sana M. Arnouk, Jan Brughmans, Eva Hadadi, Jiri Keirsse, Isaure Vanmeerbeek, Daliya Kancheva, Helena Van Damme, Máté Kiss, and Aleksandar Murgaski
- Abstract
Supplementary Figure from Efficacy of CD40 Agonists Is Mediated by Distinct cDC Subsets and Subverted by Suppressive Macrophages
- Published
- 2023
5. Supplementary Data from Efficacy of CD40 Agonists Is Mediated by Distinct cDC Subsets and Subverted by Suppressive Macrophages
- Author
-
Damya Laoui, Carola H. Ries, Martina Schmittnaegel, Greetje Vande Velde, Abhishek D. Garg, Sophie Janssens, Sofie Deschoemaeker, Niels Vandamme, Sabine Hoves, Louis Boon, Yvon Elkrim, Victor Bosteels, Ayla Debraekeleer, Ahmed E.I. Hamouda, Sana M. Arnouk, Jan Brughmans, Eva Hadadi, Jiri Keirsse, Isaure Vanmeerbeek, Daliya Kancheva, Helena Van Damme, Máté Kiss, and Aleksandar Murgaski
- Abstract
Supplementary Data from Efficacy of CD40 Agonists Is Mediated by Distinct cDC Subsets and Subverted by Suppressive Macrophages
- Published
- 2023
6. Supplementary Tables S1-S2 from M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization in the Tumor Microenvironment
- Author
-
Damya Laoui, Jo A. Van Ginderachter, Patrick De Baetselier, Frank Tacke, Lars Vereecke, Can Ergen, Qods Lahmar, Chloé Abels, Lea Brys, Yvon Elkrim, Yannick Morias, Jiri Keirsse, Felix Heymann, Benoît Stijlemans, and Eva Van Overmeire
- Abstract
Antibodies for flow cytometry (S1); List of gene specific primer sequences for qRT-PCR (S2).
- Published
- 2023
7. Data from M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization in the Tumor Microenvironment
- Author
-
Damya Laoui, Jo A. Van Ginderachter, Patrick De Baetselier, Frank Tacke, Lars Vereecke, Can Ergen, Qods Lahmar, Chloé Abels, Lea Brys, Yvon Elkrim, Yannick Morias, Jiri Keirsse, Felix Heymann, Benoît Stijlemans, and Eva Van Overmeire
- Abstract
Tumors contain a heterogeneous myeloid fraction comprised of discrete MHC-IIhi and MHC-IIlo tumor-associated macrophage (TAM) subpopulations that originate from Ly6Chi monocytes. However, the mechanisms regulating the abundance and phenotype of distinct TAM subsets remain unknown. Here, we investigated the role of macrophage colony-stimulating factor (M-CSF) in TAM differentiation and polarization in different mouse tumor models. We demonstrate that treatment of tumor-bearing mice with a blocking anti-M-CSFR monoclonal antibody resulted in a reduction of mature TAMs due to impaired recruitment, extravasation, proliferation, and maturation of their Ly6Chi monocytic precursors. M-CSFR signaling blockade shifted the MHC-IIlo/MHC-IIhi TAM balance in favor of the latter as observed by the preferential differentiation of Ly6Chi monocytes into MHC-IIhi TAMs. In addition, the genetic and functional signatures of MHC-IIlo TAMs were downregulated upon M-CSFR blockade, indicating that M-CSFR signaling shapes the MHC-IIlo TAM phenotype. Conversely, granulocyte macrophage (GM)-CSFR had no effect on the mononuclear tumor infiltrate or relative abundance of TAM subsets. However, GM-CSFR signaling played an important role in fine-tuning the MHC-IIhi phenotype. Overall, our data uncover the multifaceted and opposing roles of M-CSFR and GM-CSFR signaling in governing the phenotype of macrophage subsets in tumors, and provide new insight into the mechanism of action underlying M-CSFR blockade. Cancer Res; 76(1); 35–42. ©2015 AACR.
- Published
- 2023
8. Supplementary Figures S1-S9 from M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization in the Tumor Microenvironment
- Author
-
Damya Laoui, Jo A. Van Ginderachter, Patrick De Baetselier, Frank Tacke, Lars Vereecke, Can Ergen, Qods Lahmar, Chloé Abels, Lea Brys, Yvon Elkrim, Yannick Morias, Jiri Keirsse, Felix Heymann, Benoît Stijlemans, and Eva Van Overmeire
- Abstract
Effects of α-M-CSFR Ab treatment on Ly6Chi monocyte function (S1); Effect of M-CSFR blockade on the survival of Ly6Chi monocytes and TAM (S2); Effect of M-CSFR blockade on Ly6Chi monocyte percentages in peripheral blood, spleen and bone marrow (S3); Absolute numbers of myeloid cell subsets in tumors from isotype or α-M-CSFR Ab-treated animals (S4); Production of M-CSF, IL-34 and GM-CSF by 3LL-R tumors (S5); Effect of M-CSFR blockade on monocyte differentiation in TS/A breast carcinoma tumors (S6); M-CSFR-mediated regulation of gene expression is IL-4Rα- independent (S7); Mixed Leukocyte Reaction (MLR) by TAM subsets from isotype or α-M-CSFR Ab treated animals (S8); M-CSFR blockade enhanced the M1-like MHC-IIhi TAM gene signature independently of GM-CSFR signaling (S9).
- Published
- 2023
9. Supplementary Figures 1 through 5 from Tumor Hypoxia Does Not Drive Differentiation of Tumor-Associated Macrophages but Rather Fine-Tunes the M2-like Macrophage Population
- Author
-
Jo A. Van Ginderachter, Massimiliano Mazzone, Patrick De Baetselier, Conny Gysemans, Peter Carmeliet, Bénédicte Jordan, Oussama Karroum, Yvon Elkrim, Elio Schouppe, Isabelle Houbracken, Kiavash Movahedi, Yannick Morias, Jiri Keirsse, Chiara Aldeni, Giusy Di Conza, Eva Van Overmeire, and Damya Laoui
- Abstract
PDF - 410K, Molecular and functional heterogeneity of TAM and TADC subsets in 3LL-R tumors (S1); Characterization of the myeloid cell infiltrate in orthotopic 3LL-R tumors and effect of tumor growth on the lung macrophage subsets (S2); Intratumoral localization of the distinct myeloid cell subpopulations in 3LL-R tumors (S3); PHD2-haplodeficiency increases tumor vessel perfusion and vessel coverage in 3LL-R tumors (S4); Expression of hypoxia-regulated proteins in sorted TAM (S5).
- Published
- 2023
10. Supplementary Methods from Tumor Hypoxia Does Not Drive Differentiation of Tumor-Associated Macrophages but Rather Fine-Tunes the M2-like Macrophage Population
- Author
-
Jo A. Van Ginderachter, Massimiliano Mazzone, Patrick De Baetselier, Conny Gysemans, Peter Carmeliet, Bénédicte Jordan, Oussama Karroum, Yvon Elkrim, Elio Schouppe, Isabelle Houbracken, Kiavash Movahedi, Yannick Morias, Jiri Keirsse, Chiara Aldeni, Giusy Di Conza, Eva Van Overmeire, and Damya Laoui
- Abstract
PDF - 49K, Description of additional methods and procedures used in the study. Also includes supplementary references.
- Published
- 2023
11. Supplementary Methods from M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization in the Tumor Microenvironment
- Author
-
Damya Laoui, Jo A. Van Ginderachter, Patrick De Baetselier, Frank Tacke, Lars Vereecke, Can Ergen, Qods Lahmar, Chloé Abels, Lea Brys, Yvon Elkrim, Yannick Morias, Jiri Keirsse, Felix Heymann, Benoît Stijlemans, and Eva Van Overmeire
- Abstract
Description of additional methods and procedures used in the study.
- Published
- 2023
12. Supplementary Tables 1 and 2 from Tumor Hypoxia Does Not Drive Differentiation of Tumor-Associated Macrophages but Rather Fine-Tunes the M2-like Macrophage Population
- Author
-
Jo A. Van Ginderachter, Massimiliano Mazzone, Patrick De Baetselier, Conny Gysemans, Peter Carmeliet, Bénédicte Jordan, Oussama Karroum, Yvon Elkrim, Elio Schouppe, Isabelle Houbracken, Kiavash Movahedi, Yannick Morias, Jiri Keirsse, Chiara Aldeni, Giusy Di Conza, Eva Van Overmeire, and Damya Laoui
- Abstract
PDf - 36K, List of antibodies (S1); List of gene specific primers (S2).
- Published
- 2023
13. Peroxiredoxin-1 is an H2o2 Safe-Guard Antioxidant and Signalling Enzyme in Macrophages Independent of Their Polarization State
- Author
-
Daria Ezeriņa, Nghia Vo Trung, Ting Luo, Yvon Elkrim, Anna Escoda Suarez, Gaëtan Herinckx, Didier Vertommen, Damya Laoui, Jo A. Van Ginderachter, and Joris Messens
- Published
- 2023
14. Junctional adhesion molecule-A is dispensable for myeloid cell recruitment and diversification in the tumor microenvironment
- Author
-
Máté Kiss, Els Lebegge, Aleksandar Murgaski, Helena Van Damme, Daliya Kancheva, Jan Brughmans, Isabelle Scheyltjens, Ali Talebi, Robin Maximilian Awad, Yvon Elkrim, Pauline M. R. Bardet, Sana M. Arnouk, Cleo Goyvaerts, Johan Swinnen, Frank Aboubakar Nana, Jo A. Van Ginderachter, Damya Laoui, Brussels Heritage Lab, Cellular and Molecular Immunology, Faculty of Sciences and Bioengineering Sciences, Department of Bio-engineering Sciences, Basic (bio-) Medical Sciences, Laboratory of Molecullar and Cellular Therapy, Faculty of Medicine and Pharmacy, UCL - SSS/IREC/MONT - Pôle Mont Godinne, and UCL - (MGD) Service de pneumologie
- Subjects
Pulmonary and Respiratory Medicine ,mice ,Tumor Microenvironment/genetics ,Immunology ,F11R ,PROTECTS ,Mice ,tumor-associated macrophage ,INFLAMMATION ,Monocytes/metabolism ,Tumor Microenvironment ,Animals ,Immunology and Allergy ,Myeloid Cells ,JAM-A ,Science & Technology ,Inflammation/metabolism ,Myeloid Cells/metabolism ,ENDOTHELIAL-CELLS ,JAM-1 ,Junctional Adhesion Molecule A ,MONOCYTES ,oncology ,monocyte ,junctional adhesion molecule-A ,MACROPHAGE ,REGULATOR ,Life Sciences & Biomedicine ,extravasation ,interleukin-1 - Abstract
Junctional adhesion molecule-A (JAM-A), expressed on the surface of myeloid cells, is required for extravasation at sites of inflammation and may also modulate myeloid cell activation. Infiltration of myeloid cells is a common feature of tumors that drives disease progression, but the function of JAM-A in this phenomenon and its impact on tumor-infiltrating myeloid cells is little understood. Here we show that systemic cancer-associated inflammation in mice enhanced JAM-A expression selectively on circulating monocytes in an IL1β-dependent manner. Using myeloid-specific JAM-A-deficient mice, we found that JAM-A was dispensable for recruitment of monocytes and other myeloid cells to tumors, in contrast to its reported role in inflammation. Single-cell RNA sequencing revealed that loss of JAM-A did not influence the transcriptional reprogramming of myeloid cells in the tumor microenvironment. Overall, our results support the notion that cancer-associated inflammation can modulate the phenotype of circulating immune cells, and we demonstrate that tumors can bypass the requirement of JAM-A for myeloid cell recruitment and reprogramming. ispartof: FRONTIERS IN IMMUNOLOGY vol:13 ispartof: location:Switzerland status: published
- Published
- 2022
15. Efficacy of CD40 agonists is mediated by distinct cDC subsets and subverted by suppressive macrophages
- Author
-
Aleksandar Murgaski, Máté Kiss, Helena Van Damme, Daliya Kancheva, Isaure Vanmeerbeek, Jiri Keirsse, Eva Hadadi, Jan Brughmans, Sana M. Arnouk, Ahmed E.I. Hamouda, Ayla Debraekeleer, Victor Bosteels, Yvon Elkrim, Louis Boon, Sabine Hoves, Niels Vandamme, Sofie Deschoemaeker, Sophie Janssens, Abhishek D. Garg, Greetje Vande Velde, Martina Schmittnaegel, Carola H. Ries, Damya Laoui, Cellular and Molecular Immunology, Department of Bio-engineering Sciences, and Faculty of Sciences and Bioengineering Sciences
- Subjects
Cancer Research ,RECEPTOR ,Macrophages ,Biology and Life Sciences ,Dendritic Cells ,CD8-Positive T-Lymphocytes ,DENDRITIC CELLS ,THERAPY ,MECHANISMS ,Mice, Inbred C57BL ,Mice ,Oncology ,ANTIBODY ,LEADS ,Neoplasms ,Medicine and Health Sciences ,Animals ,Humans ,TRIAL ,HELP ,CD40 Antigens ,COMBINATION ,CP-870,893 - Abstract
Agonistic αCD40 therapy has been shown to inhibit cancer progression in only a fraction of patients. Understanding the cancer cell–intrinsic and microenvironmental determinants of αCD40 therapy response is therefore crucial to identify responsive patient populations and to design efficient combinatorial treatments. Here, we show that the therapeutic efficacy of αCD40 in subcutaneous melanoma relies on preexisting, type 1 classical dendritic cell (cDC1)–primed CD8+ T cells. However, after administration of αCD40, cDC1s were dispensable for antitumor efficacy. Instead, the abundance of activated cDCs, potentially derived from cDC2 cells, increased and further activated antitumor CD8+ T cells. Hence, distinct cDC subsets contributed to the induction of αCD40 responses. In contrast, lung carcinomas, characterized by a high abundance of macrophages, were resistant to αCD40 therapy. Combining αCD40 therapy with macrophage depletion led to tumor growth inhibition only in the presence of strong neoantigens. Accordingly, treatment with immunogenic cell death–inducing chemotherapy sensitized lung tumors to αCD40 therapy in subcutaneous and orthotopic settings. These insights into the microenvironmental regulators of response to αCD40 suggest that different tumor types would benefit from different combinations of therapies to optimize the clinical application of CD40 agonists. Significance: This work highlights the temporal roles of different dendritic cell subsets in promoting CD8+ T-cell–driven responses to CD40 agonist therapy in cancer.
- Published
- 2022
16. Intrabody Targeting HIF-1α Mediates Transcriptional Downregulation of Target Genes Related to Solid Tumors
- Author
-
Cécile Vincke, Marylène Vandevenne, Changxiao Liu, Serge Muyldermans, Yaozhong Hu, Ema Romão, Yvon Elkrim, Lea Brys, Department of Bio-engineering Sciences, Cellular and Molecular Immunology, and Faculty of Sciences and Bioengineering Sciences
- Subjects
Phage display ,Transcription, Genetic ,Uterine Cervical Neoplasms ,Down-Regulation/drug effects ,Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors ,Intrabody ,Transcription, Genetic/drug effects ,Transcription (biology) ,Transcriptional regulation ,Biology (General) ,Spectroscopy ,biology ,Chemistry ,General Medicine ,Uterine Cervical Neoplasms/genetics ,nanobodies ,Cell Hypoxia ,Computer Science Applications ,Cell biology ,Gene Expression Regulation, Neoplastic ,Female ,QH301-705.5 ,Immunoprecipitation ,Protein subunit ,HIF-1α ,Down-Regulation ,Transfection ,Catalysis ,Article ,Inorganic Chemistry ,Downregulation and upregulation ,Protein Domains ,Escherichia coli ,transcriptional activation ,Humans ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,Gene ,Cell Hypoxia/genetics ,Single-Domain Antibodies/genetics ,Organic Chemistry ,Single-Domain Antibodies ,Hypoxia-Inducible Factor 1, alpha Subunit ,target genes ,Escherichia coli/genetics ,Hela Cells ,biology.protein ,Protein Domains/genetics - Abstract
Uncontrolled growth of solid tumors will result in a hallmark hypoxic condition, whereby the key transcriptional regulator of hypoxia inducible factor-1α (HIF-1α) will be stabilized to activate the transcription of target genes that are responsible for the metabolism, proliferation, and metastasis of tumor cells. Targeting and inhibiting the transcriptional activity of HIF-1 may provide an interesting strategy for cancer therapy. In the present study, an immune library and a synthetic library were constructed for the phage display selection of Nbs against recombinant PAS B domain protein (rPasB) of HIF-1α. After panning and screening, seven different nanobodies (Nbs) were selected, of which five were confirmed via immunoprecipitation to target the native HIF-1α subunit. The inhibitory effect of the selected Nbs on HIF-1 induced activation of target genes has been evaluated after intracellular expression of these Nbs in HeLa cells. The dramatic inhibition of both intrabody formats on the expression of HIF-1-related target genes has been confirmed, which indicated the inhibitory efficacy of selected Nbs on the transcriptional activity of HIF-1.
- Published
- 2021
17. IL1β Promotes Immune Suppression in the Tumor Microenvironment Independent of the Inflammasome and Gasdermin D
- Author
-
Mate Kiss, Geert Raes, Maryse Schmoetten, Helena Van Damme, Andy Wullaert, Jiri Keirsse, Mohamed Lamkanfi, Yvon Elkrim, Pauline M. R. Bardet, Junbin Qian, Aleksandar Murgaski, Louis Boon, Diether Lambrechts, Samantha Pretto, Alain Beschin, Geert van Loo, Massimiliano Mazzone, Manuel Ehling, Jo A. Van Ginderachter, Damya Laoui, Sana M. Arnouk, Evangelia Bolli, Ayla Debraekeleer, Amelie Fossoul, Els Lebegge, Jan Brughmans, Lieselotte Vande Walle, Pedro Henrique Viana Saavedra, Cellular and Molecular Immunology, Faculty of Sciences and Bioengineering Sciences, and Department of Bio-engineering Sciences
- Subjects
0301 basic medicine ,Cancer Research ,Cytotoxic ,Inflammasomes ,Neutrophils ,Knockout ,T-Lymphocytes ,medicine.medical_treatment ,Interleukin-1beta ,Immunology ,Inflammation ,Cell Communication ,Mice ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Mediator ,Neoplasms ,Tumor-Associated Macrophages ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Secretion ,Lymphocytes ,Tumor-Infiltrating ,Disease Models, Animal ,Female ,Intracellular Signaling Peptides and Proteins ,Mice, Knockout ,Phosphate-Binding Proteins ,T-Lymphocytes, Cytotoxic ,Tumor Escape ,Tumor microenvironment ,Animal ,Chemistry ,Inflammasome ,Cell biology ,030104 developmental biology ,Cytokine ,Tumor progression ,030220 oncology & carcinogenesis ,Disease Models ,medicine.symptom ,medicine.drug - Abstract
IL1β is a central mediator of inflammation. Secretion of IL1β typically requires proteolytic maturation by the inflammasome and formation of membrane pores by gasdermin D (GSDMD). Emerging evidence suggests an important role for IL1β in promoting cancer progression in patients, but the underlying mechanisms are ill-defined. Here, we have shown a key role for IL1β in driving tumor progression in two distinct mouse tumor models. Notably, activation of the inflammasome, caspase-8, as well as the pore-forming proteins GSDMD and mixed lineage kinase domain-like protein in the host were dispensable for the release of intratumoral bioactive IL1β. Inflammasome-independent IL1β release promoted systemic neutrophil expansion and fostered accumulation of T-cell-suppressive neutrophils in the tumor. Moreover, IL1β was essential for neutrophil infiltration triggered by antiangiogenic therapy, thereby contributing to treatment-induced immunosuppression. Deletion of IL1β allowed intratumoral accumulation of CD8+ effector T cells that subsequently activated tumor-associated macrophages. Depletion of either CD8+ T cells or macrophages abolished tumor growth inhibition in IL1β-deficient mice, demonstrating a crucial role for CD8+ T-cell-macrophage cross-talk in the antitumor immune response. Overall, these results support a tumor-promoting role for IL1β through establishing an immunosuppressive microenvironment and show that inflammasome activation is not essential for release of this cytokine in tumors. ispartof: CANCER IMMUNOLOGY RESEARCH vol:9 issue:3 pages:309-323 ispartof: location:United States status: published
- Published
- 2021
18. Myeloid tumor necrosis factor and heme oxygenase-1 regulate the progression of colorectal liver metastases during hepatic ischemia-reperfusion
- Author
-
Jean-François Hastir, Sophie Laurent, P. Demetter, Desislava Germanova, Lionel Larbanoix, Sergei A. Nedospasov, Laurine Verset, Nicolas Preyat, Jiri Keirsse, Sandrine Delbauve, Yvon Elkrim, Arnaud Köhler, Jo A. Van Ginderachter, Véronique Flamand, Vincent Donckier, Cellular and Molecular Immunology, and Department of Bio-engineering Sciences
- Subjects
Male ,Cancer Research ,liver ischemia-reperfusion ,Necrosis ,Myeloid ,Kupffer Cells ,Inflammation ,Monocytes ,03 medical and health sciences ,colorectal metastasis ,Mice ,0302 clinical medicine ,Medicine ,Animals ,business.industry ,Tumor Necrosis Factor-alpha ,Monocyte ,Liver Neoplasms ,Interleukin ,Sciences bio-médicales et agricoles ,Heme oxygenase ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Liver ,Tumor progression ,inflammation ,030220 oncology & carcinogenesis ,Receptor-Interacting Protein Serine-Threonine Kinases ,Reperfusion Injury ,Cancer research ,Disease Progression ,Tumor necrosis factor alpha ,medicine.symptom ,Neoplasm Recurrence, Local ,business ,Colorectal Neoplasms ,Heme Oxygenase-1 - Abstract
The liver ischemia-reperfusion (IR) injury that occurs consequently to hepatic resection performed in patients with metastases can lead to tumor relapse for not fully understood reasons. We assessed the effects of liver IR on tumor growth and the innate immune response in a mouse model of colorectal (CR) liver metastasis. Mice subjected to liver ischemia 2 days after intrasplenic injection of CR carcinoma cells displayed a higher metastatic load in the liver, correlating with Kupffer cells (KC) death through the activation of receptor-interating protein 3 kinase (RIPK3) and caspase-1 and a recruitment of monocytes. Interestingly, the immunoregulatory mediators, tumor necrosis factor-α (TNF-α) and heme oxygenase-1 (HO-1) were strongly upregulated in recruited monocytes and were also expressed in the surviving KC following IR. Using TNFflox/flox LysMcre/wt mice, we showed that TNF deficiency in macrophages and monocytes favors tumor progression after IR. The antitumor effect of myeloid cell-derived TNF involved direct tumor cell apoptosis and a reduced expression of immunosuppressive molecules such as transforming growth factor-β, interleukin (IL)-10, inducible nitric oxyde synthase (iNOS), IL-33 and HO-1. Conversely, a monocyte/macrophage-specific deficiency in HO-1 (HO-1flox/flox LysMcre/wt ) or the blockade of HO-1 function led to the control of tumor progression post-liver IR. Importantly, host cell RIPK3 deficiency maintains the KC number upon IR, inhibits the IR-induced innate cell recruitment, increases the TNF level, decreases the HO-1 level and suppresses the tumor outgrowth. In conclusion, tumor recurrence in host undergoing liver IR is associated with the death of antitumoral KC and the recruitment of monocytes endowed with immunosuppressive properties. In both of which HO-1 inhibition would reinforce their antitumoral activity., info:eu-repo/semantics/published
- Published
- 2021
19. Therapeutic depletion of CCR8+ tumor-infiltrating regulatory T cells elicits antitumor immunity and synergizes with anti-PD-1 therapy
- Author
-
Niels Vandamme, Louis Boon, Jo A. Van Ginderachter, Maude Jans, Damya Laoui, Yvon Elkrim, Pauline M. R. Bardet, Evangelia Bolli, Mate Kiss, Bart Neyns, Aleksandar Murgaski, Elizabeth Allen, Bruno Dombrecht, Sebahat Ocak, Heleen Roose, Lars Vereecke, Frank Aboubakar Nana, Pascal Merchiers, James Dooley, Helena Van Damme, Daliya Kancheva, Maria Solange Martins, Gillian Blancke, Frank Tacke, Eva Van Overmeire, Isabelle Scheyltjens, Kiavash Movahedi, Julia Katharina Schwarze, Liesbet Martens, Cellular and Molecular Immunology, Faculty of Sciences and Bioengineering Sciences, Department of Bio-engineering Sciences, Internal Medicine, Clinical sciences, Faculty of Medicine and Pharmacy, Basic (bio-) Medical Sciences, Medical Oncology, Laboratory for Medical and Molecular Oncology, Laboratory of Molecullar and Cellular Therapy, Van Damme, Helena [0000-0002-1042-2907], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,lymphocytes ,Cancer Research ,Lung Neoplasms ,Skin Neoplasms ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Melanoma, Experimental ,receptors ,CCR8 ,T-Lymphocytes, Regulatory ,Epitope ,Carcinoma, Lewis Lung ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Immunotherapy Biomarkers ,Databases, Genetic ,Immunology and Allergy ,immunologic ,Molecular Targeted Therapy ,RNA-Seq ,Immune Checkpoint Inhibitors ,RC254-282 ,Antibody-dependent cell-mediated cytotoxicity ,Mice, Knockout ,education.field_of_study ,Melanoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,hemic and immune systems ,Combined Modality Therapy ,Phenotype ,Oncology ,030220 oncology & carcinogenesis ,oncology ,Molecular Medicine ,Female ,immunotherapy ,Life Sciences & Biomedicine ,biotechnology ,tumor ,Population ,Immunology ,chemical and pharmacologic phenomena ,Biology ,Lymphocyte Depletion ,Receptors, CCR8 ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,medicine ,Animals ,Humans ,education ,Pharmacology ,Science & Technology ,Gene Expression Profiling ,Biology and Life Sciences ,biomarkers ,Immunotherapy ,tumor-infiltrating ,medicine.disease ,Fusion protein ,Mice, Inbred C57BL ,030104 developmental biology ,Cancer research ,CD8 - Abstract
BackgroundModulation and depletion strategies of regulatory T cells (Tregs) constitute valid approaches in antitumor immunotherapy but suffer from severe adverse effects due to their lack of selectivity for the tumor-infiltrating (ti-)Treg population, indicating the need for a ti-Treg specific biomarker.MethodsWe employed single-cell RNA-sequencing in a mouse model of non-small cell lung carcinoma (NSCLC) to obtain a comprehensive overview of the tumor-infiltrating T-cell compartment, with a focus on ti-Treg subpopulations. These findings were validated by flow cytometric analysis of both mouse (LLC-OVA, MC38 and B16-OVA) and human (NSCLC and melanoma) tumor samples. We generated two CCR8-specific nanobodies (Nbs) that recognize distinct epitopes on the CCR8 extracellular domain. These Nbs were formulated as tetravalent Nb-Fc fusion proteins for optimal CCR8 binding and blocking, containing either an antibody-dependent cell-mediated cytotoxicity (ADCC)-deficient or an ADCC-prone Fc region. The therapeutic use of these Nb-Fc fusion proteins was evaluated, either as monotherapy or as combination therapy with anti-programmed cell death protein-1 (anti-PD-1), in both the LLC-OVA and MC38 mouse models.ResultsWe were able to discern two ti-Treg populations, one of which is characterized by the unique expression of Ccr8 in conjunction with Treg activation markers. Ccr8 is also expressed by dysfunctional CD4+ and CD8+ T cells, but the CCR8 protein was only prominent on the highly activated and strongly T-cell suppressive ti-Treg subpopulation of mouse and human tumors, with no major CCR8-positivity found on peripheral Tregs. CCR8 expression resulted from TCR-mediated Treg triggering in an NF-κB-dependent fashion, but was not essential for the recruitment, activation nor suppressive capacity of these cells. While treatment of tumor-bearing mice with a blocking ADCC-deficient Nb-Fc did not influence tumor growth, ADCC-prone Nb-Fc elicited antitumor immunity and reduced tumor growth in synergy with anti-PD-1 therapy. Importantly, ADCC-prone Nb-Fc specifically depleted ti-Tregs in a natural killer (NK) cell-dependent fashion without affecting peripheral Tregs.ConclusionsCollectively, our findings highlight the efficacy and safety of targeting CCR8 for the depletion of tumor-promoting ti-Tregs in combination with anti-PD-1 therapy.
- Published
- 2021
20. Inflammasome- and gasdermin D-independent IL-1β production mobilizes neutrophils to inhibit antitumor immunity
- Author
-
Van Ginderachter Ja, Samantha Pretto, Amelie Fossoul, Mohamed Lamkanfi, Manuel Ehling, Mate Kiss, Damya Laoui, Martins Ms, Andy Wullaert, Yvon Elkrim, Aleksandar Murgaski, Jiri Keirsse, Els Lebegge, D Lambrechts, Massimiliano Mazzone, Vande Walle L, Junbin Qian, Van Damme H, and Evangelia Bolli
- Subjects
Mediator ,Immune system ,Effector ,Tumor progression ,Chemistry ,medicine ,Cancer research ,Cytotoxic T cell ,Inflammasome ,Inflammation ,Secretion ,medicine.symptom ,medicine.drug - Abstract
Interleukin-1β (IL-1β) is a central mediator of inflammation whose secretion typically requires proteolytic maturation by the inflammasome and formation of membrane pores by gasdermin D (GSDMD). Emerging evidence suggests an important role for IL-1β in promoting cancer progression in patients, but the underlying mechanisms are little understood. Here, we show a key role for IL-1β in driving tumor progression in two distinct mouse tumor models. Notably, inflammasome activation and GSDMD were dispensable for the production of intratumoral bioactive IL-1β, which promoted systemic mobilization and infiltration of neutrophils into tumors. Neutrophils recruited via IL-1β suppressed the acquisition of an effector T-cell phenotype and subsequent antitumor immune response. Moreover, IL-1β was essential for neutrophil accumulation upon antiangiogenic therapy, thereby contributing to therapy-induced immunosuppression. Antitumor immunity in the absence of IL-1β-dependent neutrophil recruitment relied on immunostimulatory macrophages which promoted the infiltration and activation of cytotoxic T-cells. Overall, these results support a tumor-promoting role for IL-1β through establishing an immunosuppressive microenvironment and show that inflammasome activation is not essential for its release in tumors.
- Published
- 2020
21. Macrophages are Metabolically Heterogeneous within the Tumor Microenvironment
- Author
-
Xenia Geeraerts, Emile J. Clappaert, Aleksandar Murgaski, Sarah-Maria Fendt, Liesbet Martens, Yvan Saeys, Yvon Elkrim, Dorien Broekaert, Jan Van den Bossche, Kyra E. de Goede, Damya Laoui, Pauline M. R. Bardet, Jo A. Van Ginderachter, Conny Gysemans, and Juan Fernández Garcia
- Subjects
Transcriptome ,Citric acid cycle ,Tumor microenvironment ,Metabolomics ,stomatognathic system ,Tumor progression ,Chemistry ,Macrophage ,Glycolysis ,Metabolism ,skin and connective tissue diseases ,hormones, hormone substitutes, and hormone antagonists ,Cell biology - Abstract
Macrophages are often prominently present in the tumor microenvironment, where distinct macrophage populations can differentially affect tumor progression. Although the metabolism of macrophages influences their function, little is known about the metabolic characteristics of tumor-associated macrophage (TAM) subsets. Using transcriptomic and metabolomic analyses, we now reveal that pro-inflammatory MHC-IIhi TAMs display a hampered TCA cycle, while reparative MHC-IIlo TAMs show a higher oxidative and glycolytic metabolism. Both TAM populations preferred lactate over glucose as a carbon source to fuel the TCA cycle. However, while lactate supported the oxidative metabolism in MHC-IIlo TAMs, it decreased the metabolic activity of MHC-IIhi TAMs. Lactate subtly affected the transcriptome of MHC-IIlo TAMs, increased L-arginine metabolism and enhanced T-cell suppressive capacity of these TAMs. Overall, our data uncover the metabolic intricacies of distinct TAM subsets and identify lactate as an important carbon source, and metabolic and functional regulator of TAMs.
- Published
- 2020
22. STAT6 mediates footpad immunopathology in the absence of IL-12p40 following infection of susceptible BALB/c mice with Leishmania major
- Author
-
Kaat de Jonge, Olivier Denis, Yvon Elkrim, F Kauffmann, Elyn Meert, Carl De Trez, Stefan Magez, Eric Muraille, Delphine Hanot Mambres, Faculty of Sciences and Bioengineering Sciences, Cellular and Molecular Immunology, Faculty of Economic and Social Sciences and Solvay Business School, and Department of Bio-engineering Sciences
- Subjects
0301 basic medicine ,CUTANEOUS LEISHMANIASIS ,Neutrophils ,medicine.medical_treatment ,STAT6 ,neutrophils ,Medicine and Health Sciences ,BALB/c mice ,Immunology and Allergy ,immunopathology ,Leishmania major ,STAT6−/− ,IN-VIVO ,IL-12p40 ,Original Research ,Mice, Knockout ,Mice, Inbred BALB C ,biology ,Interleukin-12 Subunit p40 ,IMMUNE-RESPONSES ,Interleukin ,IL-12p40(-/-) ,IL-12p40−/− ,APOPTOSIS ,Cytokine ,Interleukin 12 ,RESISTANT MICE ,lcsh:Immunologic diseases. Allergy ,Immunology ,Leishmaniasis, Cutaneous ,Immunopathology ,BALB/c ,03 medical and health sciences ,Cutaneous leishmaniasis ,REVEALS ,medicine ,Animals ,Foot ,Intracellular parasite ,Biology and Life Sciences ,Généralités ,STAT6(-/-) ,biology.organism_classification ,medicine.disease ,INTERLEUKIN-12 ,030104 developmental biology ,RESOLUTION ,STAT6 Transcription Factor ,lcsh:RC581-607 ,SKIN - Abstract
Leishmania major (L. major) parasites are intracellular parasites belong to the Trypanosomatidae family and are the causative agent of cutaneous leishmaniasis. This disease affects approximately 1.5 million per year worldwide and there is currently no prophylactic vaccine available. L. major is transmitted by the bite of an infected sandfly and has been considered for decades now as a mouse model of choice to identify the factors implicated in T helper (Th)1 and Th2 polarization due to the natural resistance and susceptibility to infection of C57BL/6 and BALB/c mice, respectively. In this study, we refine the role of IL-12p40 cytokine, which is implicated the development of a protective Th1 response, and STAT6, a transcription factor involved in the signaling via detrimental interleukin (IL)-4 and IL-13 associated Th2 cytokines during L. major infection in the BALB/c model. In the absence of STAT6 and IL-12p40 signaling, double knockout (DKO) susceptible BALB/c mice displayed reduced footpad swelling and ulcerative lesion compared to IL-12p40-/- mice upon L. major infection. Hence, they expressed slower upregulation of keratinocyte markers implicated in the inhibition of wound healing, such as keratin 6a (Krt6a) and Krt16. This coincides with the presence of neutrophils displaying an altered phenotype characterized by a lower expression of surface markers Ly6C, CD11b, and Ly6G. These neutrophils exhibited very lower levels of apoptosis similarly to neutrophils present in resistant STAT6-/- mice. Interestingly, the reduced footpad swelling in DKO mice is associated with a high footpad parasite level similar to susceptible IL-12p40-/- mice. In conclusion, this study demonstrate that in the absence of both STAT6 and IL-12p40 signaling, L. major-infected mice display smaller and less ulcerated lesions, which does, however, not correlate with reduced parasite load. In addition, the presence of neutrophils with an altered phenotype is associated with reduced apoptosis and delayed immunopathologies, demonstrating the detrimental role of STAT6 in infected susceptible BALB/c mice., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2018
23. Macrophage dynamics are regulated by local macrophage proliferation and monocyte recruitment in injured pancreas
- Author
-
Nico De Leu, Mark Van de Casteele, Benoit Stijlemans, Patrick De Baetselier, Eva Van Overmeire, Jo A. Van Ginderachter, Harry Heimberg, Yves Heremans, Ying Cai, Conny Gysemans, Naomi Van Gassen, Sofie De Groef, Yvon Elkrim, and Gunter Leuckx
- Subjects
biology ,Cell growth ,Monocyte ,Immunology ,Cell ,biology.organism_classification ,Neogenesis ,Cell biology ,Chemokine receptor ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Beta cell ,Receptor ,Macrophage proliferation - Abstract
Pancreas injury by partial duct ligation (PDL) activates a healing response, encompassing β-cell neogenesis and proliferation. Macrophages (MΦs) were recently shown to promote β-cell proliferation after PDL, but they remain poorly characterized. We assessed myeloid cell diversity and the factors driving myeloid cell dynamics following acute pancreas injury by PDL. In naive and sham-operated pancreas, the myeloid cell compartment consisted mainly of two distinct tissue-resident MΦ types, designated MHC-II(lo) and MHC-II(hi) MΦs, the latter being predominant. MHC-II(lo) and MHC-II(hi) pancreas MΦs differed at the molecular level, with MHC-II(lo) MΦs being more M2-activated. After PDL, there was an early surge of Ly6C(hi) monocyte infiltration in the pancreas, followed by a transient MHC-II(lo) MΦ peak and ultimately a restoration of the MHC-II(hi) MΦ-dominated steady-state equilibrium. These intricate MΦ dynamics in PDL pancreas depended on monocyte recruitment by C-C chemokine receptor 2 and macrophage-colony stimulating factor receptor as well as on macrophage-colony stimulating factor receptor-dependent local MΦ proliferation. Functionally, MHC-II(lo) MΦs were more angiogenic. We further demonstrated that, at least in C-C chemokine receptor 2-KO mice, tissue MΦs, rather than Ly6C(hi) monocyte-derived MΦs, contributed to β-cell proliferation. Together, our study fully characterizes the MΦ subsets in the pancreas and clarifies the complex dynamics of MΦs after PDL injury.
- Published
- 2015
24. CCR2-dependent monocyte-derived macrophages resolve inflammation and restore gut motility in postoperative ileus
- Author
-
Jo A. Van Ginderachter, Michelle Stakenborg, Gianluca Matteoli, Pedro J. Gomez-Pinilla, Evelien Labeeuw, Damya Laoui, Elisa Meroni, Kristin A. Sauter, Martina Di Giovangiulio, Francesca D'Errico, Gera Goverse, Yvon Elkrim, Giovanna Farro, Zofia M. Lisowski, Nathalie Stakenborg, David A. Hume, Guy E. Boeckxstaens, Department of Bio-engineering Sciences, and Cellular and Molecular Immunology
- Subjects
0301 basic medicine ,Macrophage colony-stimulating factor ,CCR2 ,mice ,Receptors, CCR2 ,Motility ,Inflammation ,Macrophages/immunology ,gastrointestinal motility ,Monocytes ,03 medical and health sciences ,Chemokine receptor ,Ileus/immunology ,0302 clinical medicine ,Ileus ,Postoperative Complications ,Cell Movement ,Immunopathology ,Medicine ,Homeostasis ,Animals ,Gastrointestinal Transit ,Receptors, CCR2/immunology ,business.industry ,Monocyte ,Macrophages ,Gastroenterology ,Monocytes/immunology ,Muscle, Smooth ,Cell Differentiation ,Inflammation/immunology ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Muscle, Smooth/pathology ,Immunology ,Homeostasis/immunology ,030211 gastroenterology & hepatology ,Bone marrow ,medicine.symptom ,Postoperative Complications/immunology ,business - Abstract
ObjectivePostoperative ileus (POI) is assumed to result from myeloid cells infiltrating the intestinalmuscularis externa(ME) in patients undergoing abdominal surgery. In the current study, we investigated the role of infiltrating monocytes in a murine model of intestinal manipulation (IM)-induced POI in order to clarify whether monocytes mediate tissue damage and intestinal dysfunction or they are rather involved in the recovery of gastrointestinal (GI) motility.DesignIM was performed in mice with defective monocyte migration to tissues (C-C motif chemokine receptor 2,Ccr2−/−mice) and wild-type (WT) mice to study the role of monocytes and monocyte-derived macrophages (MΦs) during onset and resolution of ME inflammation.ResultsAt early time points, IM-induced GI transit delay and inflammation were equal in WT andCcr2−/−mice. However, GI transit recovery after IM was significantly delayed inCcr2−/−mice compared with WT mice, associated with increased neutrophil-mediated immunopathology and persistent impaired neuromuscular function. During recovery, monocyte-derived MΦs acquire pro-resolving features that aided in the resolution of inflammation. In line, bone marrow reconstitution and treatment with MΦ colony-stimulating factor 1 enhanced monocyte recruitment and MΦ differentiation and ameliorated GI transit inCcr2−/−mice.ConclusionOur study reveals a critical role for monocyte-derived MΦs in restoring intestinal homeostasis after surgical trauma. From a therapeutic point of view, our data indicate that inappropriate targeting of monocytes may increase neutrophil-mediated immunopathology and prolong the clinical outcome of POI, while future therapies should be aimed at enhancing MΦ physiological repair functions.
- Published
- 2017
25. Abstract A083: Inflammasome-independent IL-1β release by myeloid cells promotes vessel destabilization and immune suppression in the tumor microenvironment
- Author
-
Lieselotte Vande Walle, Jiri Keirsse, Maria Solange Martins, Mohamed Lamkanfi, Jens Serneels, Helena Van Damme, Evangelia Bolli, Damya Laoui, Massimiliano Mazzone, Yvon Elkrim, Mate Kiss, Amelie Fossoul, Jo A. Van Ginderachter, and Aleksandar Murgaski
- Subjects
Cancer Research ,Tumor microenvironment ,Chemistry ,Necroptosis ,medicine.medical_treatment ,Immunology ,Pyroptosis ,Inflammasome ,Immune system ,Cytokine ,Cancer immunotherapy ,Tumor progression ,Cancer research ,medicine ,medicine.drug - Abstract
Background: Chronic inflammation in the tumor microenvironment (TME) sustained by immune cells has a crucial role both in tumor initiation and progression. One of the central cytokines of inflammation, IL-1β, is produced as a biologically inactive precursor that requires proteolytic processing by caspase-1. Activation of caspase-1 is triggered by the formation of inflammasomes, multiprotein complexes that detect microbial and endogenous danger signals primarily via NOD-like receptors, such as NLRP3 and NLRC4. Biologically active IL-1β is believed to be released through membrane pores formed by gasdermin D during a lytic form of cell death called pyroptosis. Although IL-1β-mediated inflammation has been shown to have a detrimental role in tumor progression, the signaling pathway controlling IL-1β release in the TME and the exact effect of the cytokine on antitumor T-cell responses have not been fully elucidated. A better understanding of how IL-1β release is controlled in tumors will also pave the way towards the therapeutic utilization of small-molecule inhibitors available to target NOD-like receptors and caspase-1. Methods: First, we characterized the impact of IL-1β in the TME by assessing the immune cell composition and vasculature of Lewis lung carcinomas (LLC) and E0771 breast carcinomas in IL-1β-deficient mice using flow cytometry and histologic analysis. Next, we used mice deficient in different inflammasome components, including NLRP3, NLRC4 and caspase-1, to investigate the involvement of these proteins in controlling IL-1β release in LLC and E0771 tumors. Using immunoblots and small-molecule inhibitors, we further characterized the activation of alternative enzymatic pathways and their involvement in IL-1β release by tumor-associated myeloid cells. Finally, we examined the role of pyroptosis and necroptosis in IL-1β release using gasdermin D- and MLKL-deficient mice, respectively. Release of IL-1β was assessed using ELISA and immunoblots. Results: We found that IL-1β secretion was restricted to myeloid cells and promoted tumor progression in mouse models of lung and breast carcinoma. IL-1β deletion abrogated the tumor-induced mobilization of immunosuppressive neutrophils and normalized the tumor vasculature, thereby alleviating hypoxia. Consequently, proliferation of effector T-cells in the TME was enhanced, leading to higher CD4+ and CD8+ T-cell abundance in the absence of IL-1β. We observed that, although the NLRP3 inflammasome was active in tumor-infiltrating myeloid cells, NLRP3 and caspase-1 were not essential for the proteolytic maturation of pro-IL-1β and secretion of biologically active IL-1β in the TME. Inhibition or genetic deletion of caspase-8 reduced inflammasome-independent IL-1β release, indicating that caspase-8 provides an alternative pathway for proteolytic activation and secretion of IL-1β in tumor-infiltrating myeloid cells. Moreover, IL-1β release by tumor-infiltrating myeloid cells was independent of lytic cell death modalities including gasdermin D-mediated pyroptosis and MLKL-mediated necroptosis, suggesting an alternative release mechanism for the cytokine in the TME. Conclusions: Overall, our results demonstrate that tumor-infiltrating myeloid cells are able to release IL-1β independently of inflammasomes. We show that proteolytic maturation of IL-1β via caspase-8 in myeloid cells acts as an important driver of immune suppression in the TME through vascular destabilization, recruitment of immunosuppressive neutrophils and consequential inhibition of antitumor T-cell responses. We also show, that, unlike in autoinflammation, gasdermin D-mediated pyroptosis is not essential for the release of IL-1β in tumors. These results suggest that therapeutic inhibition of inflammasomes or pyroptosis will likely not be beneficial in certain tumor types due to the presence of an alternative caspase-8-mediated IL-1β release pathway in tumor-associated myeloid cells. Citation Format: Máté Kiss, Lieselotte Vande Walle, Helena Van Damme, Aleksandar Murgaski, Evangelia Bolli, Jiri Keirsse, Maria Solange Martins, Yvon Elkrim, Amelie Fossoul, Jens Serneels, Massimiliano Mazzone, Mohamed Lamkanfi, Jo A. Van Ginderachter, Damya Laoui. Inflammasome-independent IL-1β release by myeloid cells promotes vessel destabilization and immune suppression in the tumor microenvironment [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A083.
- Published
- 2019
26. The tumour microenvironment harbours ontogenically distinct dendritic cell populations with opposing effects on tumour immunity
- Author
-
Massimiliano Mazzone, Louis Boon, Charlotte L. Scott, Martin Guilliams, Jens Serneels, Xenia Geeraerts, Yannick Morias, Jiri Keirsse, Dorine Sichien, Jo A. Van Ginderachter, Damya Laoui, Patrick De Baetselier, Qods Lahmar, Eva Van Overmeire, Yvon Elkrim, Mate Kiss, Evangelia Bolli, Department of Bio-engineering Sciences, Cellular and Molecular Immunology, Faculty of Sciences and Bioengineering Sciences, and Vriendenkring VUB
- Subjects
0301 basic medicine ,SUBSETS ,medicine.medical_treatment ,General Physics and Astronomy ,Inbred C57BL ,Transgenic ,Monocytes ,Mice ,Cancer immunotherapy ,T-Lymphocyte Subsets ,Neoplasms ,Medicine and Health Sciences ,Tumor Microenvironment ,MACROPHAGES ,Lymph node ,IN-VIVO ,Inbred BALB C ,Cells, Cultured ,Mice, Knockout ,education.field_of_study ,Mice, Inbred BALB C ,Multidisciplinary ,Tumor ,Cultured ,CYTOKINE GM-CSF ,INTRATUMORAL DELIVERY ,Multidisciplinary Sciences ,medicine.anatomical_structure ,DIFFERENTIATION ,Science & Technology - Other Topics ,CANCER-IMMUNOTHERAPY ,Immunotherapy ,Reprogramming ,STEADY-STATE ,Science ,Cells ,Knockout ,Population ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,education ,Tumor microenvironment ,Science & Technology ,Macrophages ,Dendritic Cells ,Mice, Inbred C57BL ,Biology and Life Sciences ,General Chemistry ,Dendritic cell ,030104 developmental biology ,Immunology ,Cancer research ,T-CELLS ,CD8 ,RESPONSES - Abstract
Various steady state and inflamed tissues have been shown to contain a heterogeneous DC population consisting of developmentally distinct subsets, including cDC1s, cDC2s and monocyte-derived DCs, displaying differential functional specializations. The identification of functionally distinct tumour-associated DC (TADC) subpopulations could prove essential for the understanding of basic TADC biology and for envisaging targeted immunotherapies. We demonstrate that multiple mouse tumours as well as human tumours harbour ontogenically discrete TADC subsets. Monocyte-derived TADCs are prominent in tumour antigen uptake, but lack strong T-cell stimulatory capacity due to NO-mediated immunosuppression. Pre-cDC-derived TADCs have lymph node migratory potential, whereby cDC1s efficiently activate CD8+ T cells and cDC2s induce Th17 cells. Mice vaccinated with cDC2s displayed a reduced tumour growth accompanied by a reprogramming of pro-tumoural TAMs and a reduction of MDSCs, while cDC1 vaccination strongly induces anti-tumour CTLs. Our data might prove important for therapeutic interventions targeted at specific TADC subsets or their precursors., Dendritic cells are antigen-presenting cells consisting of distinct subsets originating from different lineages. Here, the authors identify the subsets of dendritic cells populating the tumour tissue in both mice and humans and find they have opposing functions in regulating the anti-tumour immune response.
- Published
- 2016
27. M-CSF and GM-CSF receptor signaling differentially regulate monocyte maturation and macrophage polarization in the tumor microenvironment
- Author
-
Qods Lahmar, Yvon Elkrim, Yannick Morias, Frank Tacke, Felix Heymann, Eva Van Overmeire, Lars Vereecke, Damya Laoui, Jiri Keirsse, Can Ergen, Patrick De Baetselier, Jo A. Van Ginderachter, Benoit Stijlemans, Lea Brys, Chloé Abels, Department of Bio-engineering Sciences, Cellular and Molecular Immunology, and Faculty of Sciences and Bioengineering Sciences
- Subjects
0301 basic medicine ,Macrophage colony-stimulating factor ,Cancer Research ,Cellular differentiation ,Macrophage polarization ,Biology ,Monocytes ,Carcinoma, Lewis Lung ,Mice ,03 medical and health sciences ,stomatognathic system ,Receptors, Colony-Stimulating Factor ,Tumor Microenvironment ,Animals ,Macrophage ,skin and connective tissue diseases ,Tumor microenvironment ,Macrophage Colony-Stimulating Factor ,Macrophages ,GM-CSF Receptor ,Antibodies, Monoclonal ,Cell Polarity ,Mammary Neoplasms, Experimental ,Cell Differentiation ,Colony-stimulating factor ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Oncology ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Female ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
Tumors contain a heterogeneous myeloid fraction comprised of discrete MHC-IIhi and MHC-IIlo tumor-associated macrophage (TAM) subpopulations that originate from Ly6Chi monocytes. However, the mechanisms regulating the abundance and phenotype of distinct TAM subsets remain unknown. Here, we investigated the role of macrophage colony-stimulating factor (M-CSF) in TAM differentiation and polarization in different mouse tumor models. We demonstrate that treatment of tumor-bearing mice with a blocking anti-M-CSFR monoclonal antibody resulted in a reduction of mature TAMs due to impaired recruitment, extravasation, proliferation, and maturation of their Ly6Chi monocytic precursors. M-CSFR signaling blockade shifted the MHC-IIlo/MHC-IIhi TAM balance in favor of the latter as observed by the preferential differentiation of Ly6Chi monocytes into MHC-IIhi TAMs. In addition, the genetic and functional signatures of MHC-IIlo TAMs were downregulated upon M-CSFR blockade, indicating that M-CSFR signaling shapes the MHC-IIlo TAM phenotype. Conversely, granulocyte macrophage (GM)-CSFR had no effect on the mononuclear tumor infiltrate or relative abundance of TAM subsets. However, GM-CSFR signaling played an important role in fine-tuning the MHC-IIhi phenotype. Overall, our data uncover the multifaceted and opposing roles of M-CSFR and GM-CSFR signaling in governing the phenotype of macrophage subsets in tumors, and provide new insight into the mechanism of action underlying M-CSFR blockade. Cancer Res; 76(1); 35–42. ©2015 AACR.
- Published
- 2016
28. Macrophage dynamics are regulated by local macrophage proliferation and monocyte recruitment in injured pancreas
- Author
-
Naomi, Van Gassen, Eva, Van Overmeire, Gunter, Leuckx, Yves, Heremans, Sofie, De Groef, Ying, Cai, Yvon, Elkrim, Conny, Gysemans, Benoît, Stijlemans, Mark, Van de Casteele, Patrick, De Baetselier, Nico, De Leu, Harry, Heimberg, and Jo A, Van Ginderachter
- Subjects
Male ,Mice, Knockout ,Macrophages ,Histocompatibility Antigens Class II ,Pancreatic Ducts ,Mice, Transgenic ,Receptor, Macrophage Colony-Stimulating Factor ,Macrophage Activation ,Monocytes ,Mice, Inbred C57BL ,Mice ,Cellular Microenvironment ,Cell Movement ,Animals ,Antigens, Ly ,Regeneration ,Myeloid Cells ,Ligation ,Pancreas ,Cell Proliferation - Abstract
Pancreas injury by partial duct ligation (PDL) activates a healing response, encompassing β-cell neogenesis and proliferation. Macrophages (MΦs) were recently shown to promote β-cell proliferation after PDL, but they remain poorly characterized. We assessed myeloid cell diversity and the factors driving myeloid cell dynamics following acute pancreas injury by PDL. In naive and sham-operated pancreas, the myeloid cell compartment consisted mainly of two distinct tissue-resident MΦ types, designated MHC-II(lo) and MHC-II(hi) MΦs, the latter being predominant. MHC-II(lo) and MHC-II(hi) pancreas MΦs differed at the molecular level, with MHC-II(lo) MΦs being more M2-activated. After PDL, there was an early surge of Ly6C(hi) monocyte infiltration in the pancreas, followed by a transient MHC-II(lo) MΦ peak and ultimately a restoration of the MHC-II(hi) MΦ-dominated steady-state equilibrium. These intricate MΦ dynamics in PDL pancreas depended on monocyte recruitment by C-C chemokine receptor 2 and macrophage-colony stimulating factor receptor as well as on macrophage-colony stimulating factor receptor-dependent local MΦ proliferation. Functionally, MHC-II(lo) MΦs were more angiogenic. We further demonstrated that, at least in C-C chemokine receptor 2-KO mice, tissue MΦs, rather than Ly6C(hi) monocyte-derived MΦs, contributed to β-cell proliferation. Together, our study fully characterizes the MΦ subsets in the pancreas and clarifies the complex dynamics of MΦs after PDL injury.
- Published
- 2014
29. Adenosine 5'-monophosphate challenge elicits a more peripheral airway response than methacholine challenge
- Author
-
Amaryllis Haccuria, Alain Michils, Yvon Elkrim, and Alain Van Muylem
- Subjects
Adenosine monophosphate ,Adult ,Male ,Physiology ,chemistry.chemical_compound ,Physiology (medical) ,medicine ,Humans ,Respiratory system ,Lung ,Methacholine Chloride ,Asthma ,business.industry ,medicine.disease ,Adenosine ,Adenosine Monophosphate ,Methacholine challenge ,Respiratory Function Tests ,medicine.anatomical_structure ,chemistry ,Anesthesia ,Immunology ,Bronchoconstriction ,Methacholine ,Female ,medicine.symptom ,business ,Pulmonary Ventilation ,medicine.drug ,Respiratory tract - Abstract
Adenosine 5′-monophosphate (AMP) and methacholine are commonly used to assess airway hyperreactivity. However, it is not fully known whether the site of airway constriction primarily involved during challenges with either agent is similar. Using a ventilation distribution test, we investigated whether the constriction induced by each agent involves the lung periphery in a similar fashion. Ventilation distribution was evaluated by the phase III slope (S) of the single-breath washout, using gases with different diffusivities like helium (He) and hexafluorosulfur (SF6). A greater postchallenge increase in SHe reflects alterations at the level of terminal and respiratory bronchioles, while a greater increase in SSF6 reflects alterations in alveolar ducts, increases to an equal extent reflecting alterations in more proximal airways where gas transport is still convective for both gases. SSF6 and SHe were measured in 15 asthma patients before and after airway challenges (20% forced expired volume in 1-s fall) with AMP and methacholine. SHe increased to a greater extent than SSF6 after AMP challenge (5.7 vs. 3.7%/l; P = 0.002), with both slopes increasing to an equal extent after methacholine challenge (3.1%/l; P = 0.959). The larger increase in SHe following AMP challenge suggests distal ventilation impairment up to the level of terminal and respiratory bronchioles. With methacholine, the similar increases in SHe and SSF6 suggest a less distal impairment. AMP, therefore, seems to affect more extensively the very peripheral airways, whereas methacholine seems to have an effect on less distal airways.
- Published
- 2011
30. Tu1842 Intestinal Manipulation-Induced Monocyte and Neutrophil Recruitment Into the Muscularis Externa Contributes to Restore Gut Motility
- Author
-
Jo A. Van Ginderachter, Giovanna Farro, Guy E. Boeckxstaens, Gianluca Matteoli, Nathalie Stakenborg, Pedro J. Gomez-Pinilla, Martina Di Giovangiulio, Damya Laoui, and Yvon Elkrim
- Subjects
medicine.anatomical_structure ,Hepatology ,Gut motility ,Monocyte ,Immunology ,Gastroenterology ,medicine ,Biology ,Neutrophil recruitment - Published
- 2015
31. Tumor-derived lactate regulates TAM metabolism and function
- Author
-
Xenia Geeraerts, Emile Clappaert, Dorien Broekaert, Sweta Parik, Yvon Elkrim, Damya Laoui, Jan Van Den Bossche, Sarah-Maria Fendt, and Jo Van Ginderachter
32. Murine Liver Myeloid Cell Isolation Protocol
- Author
-
Alain Beschin, Benoit Stijlemans, Chloé Abels, Lea Brys, Jiri Keirsse, Amanda Sparkes, Yvon Elkrim, Jo A. Van Ginderachter, Patrick De Baetselier, Department of Bio-engineering Sciences, Cellular and Molecular Immunology, and Faculty of Sciences and Bioengineering Sciences
- Subjects
Liver injury ,Myeloid ,business.industry ,Strategy and Management ,Mechanical Engineering ,Metals and Alloys ,medicine.disease ,Industrial and Manufacturing Engineering ,medicine.anatomical_structure ,Monocyte-Derived Macrophages ,medicine ,Cancer research ,Cell isolation ,Murine liver ,business - Abstract
In homeostasis, the liver is critical for the metabolism of nutrients including sugars, lipids, proteins and iron, for the clearance of toxins, and to induce immune tolerance to gut-derived antigens. These functions predispose the liver to infection by blood-borne pathogens, and to a variety of diseases ranging from toxin and medication-induced disorders (CCl4, acetaminophen) to metabolic disorders (steatohepatitis, alcoholic liver disease, biliary obstruction, cholestasis) or autoimmunity. Chronic liver injury often progresses to life threatening fibrosis and can end in liver cirrhosis and hepatocellular carcinoma (Pellicoro et al., 2014). The liver contains parenchymal cells or hepatocytes that make up the majority of hepatic cells. It also contains non-parenchymal structural cells such as sinusoidal endothelial cells and a large number of non-parenchymal innate immune cells, mainly monocytes, neutrophils, macrophages, DCs, NK and NKT cells that can trigger an adaptive immune response in the case of infections or other pathogenic insults (Jenne and Kubes, 2013). How this immune response is regulated determines the extent of acute and chronic liver injury (Stijlemans et al., 2014). In this context, liver macrophages have been demonstrated to play central but divergent (from initiating to resolving) functions in liver injury (Sica et al., 2014). It has become clear in the last years that hepatic macrophages consist of two classes, tissue-resident macrophages, the Kupffer cells (KCs) originating from yolk sac/fetal liver progenitors and tissue-infiltrating macrophages originating from bone marrow-derived Ly6CHi monocytes (Jinhoux and Jung, 2014; Tacke and Zimmerman, 2014). Distinguishing the activities of KCs from those of monocyte-derived macrophages during liver injury or repair is currently a frontline research topic in the macrophage field. Indeed, considering that clinical management of liver failure remains problematic, a better understanding of the immune mechanisms regulating liver injury is expected to allow the development of new therapeutic modalities. Here, we describe an isolation technique for liver non-parenchymal polymorphonuclear (PMN) and mononuclear myeloid cells permitting their molecular and functional characterization.
33. Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population
- Author
-
Giusy Di Conza, Damya Laoui, Kiavash Movahedi, Isabelle Houbracken, Oussama Karroum, Yannick Morias, Patrick De Baetselier, Peter Carmeliet, Eva Van Overmeire, Massimiliano Mazzone, Elio Schouppe, Bénédicte F. Jordan, Jo A. Van Ginderachter, Jiri Keirsse, Chiara Aldeni, Conny Gysemans, Yvon Elkrim, Department of Bio-engineering Sciences, Cellular and Molecular Immunology, Pathological Anatomy, Basic (bio-) Medical Sciences, Cell Differentiation, and UCL - SSS/LDRI - Louvain Drug Research Institute
- Subjects
Cancer Research ,Myeloid ,Animals ,Cell Differentiation ,Cell Hypoxia ,Disease Models, Animal ,Female ,Macrophages ,Mice ,Mice, Inbred C57BL ,Mice, Transgenic ,Neoplasms ,Transcriptome ,hypoxia-sensitive genes ,Biology ,Inbred C57BL ,Transgenic ,stomatognathic system ,In vivo ,Gene expression ,medicine ,skin and connective tissue diseases ,Tumor hypoxia ,Animal ,Hypoxia (medical) ,Phenotype ,PHD2+/+ → PHD2+/− ,medicine.anatomical_structure ,Oncology ,Integrin alpha M ,Disease Models ,Cancer cell ,Immunology ,Cancer research ,biology.protein ,prolyl hydroxylase domain 2 (PHD2) ,medicine.symptom ,MHC-IIlo MHC-IIhi ,hormones, hormone substitutes, and hormone antagonists - Abstract
Tumor-associated macrophages (TAM) are exposed to multiple microenvironmental cues in tumors, which collaborate to endow these cells with protumoral activities. Hypoxia, caused by an imbalance in oxygen supply and demand because of a poorly organized vasculature, is often a prominent feature in solid tumors. However, to what extent tumor hypoxia regulates the TAM phenotype in vivo is unknown. Here, we show that the myeloid infiltrate in mouse lung carcinoma tumors encompasses two morphologically distinct CD11bhiF4/80hiLy6Clo TAM subsets, designated as MHC-IIlo and MHC-IIhi TAM, both of which were derived from tumor-infiltrating Ly6Chi monocytes. MHC-IIlo TAM express higher levels of prototypical M2 markers and reside in more hypoxic regions. Consequently, MHC-IIlo TAM contain higher mRNA levels for hypoxia-regulated genes than their MHC-IIhi counterparts. To assess the in vivo role of hypoxia on these TAM features, cancer cells were inoculated in prolyl hydroxylase domain 2 (PHD2)-haplodeficient mice, resulting in better-oxygenated tumors. Interestingly, reduced tumor hypoxia did not alter the relative abundance of TAM subsets nor their M2 marker expression, but specifically lowered hypoxia-sensitive gene expression and angiogenic activity in the MHC-IIlo TAM subset. The same observation in PHD2+/+→ PHD2+/− bone marrow chimeras also suggests organization of a better-oxygenized microenvironment. Together, our results show that hypoxia is not a major driver of TAM subset differentiation, but rather specifically fine-tunes the phenotype of M2-like MHC-IIlo TAM. Cancer Res; 74(1); 24–30. ©2013 AACR.
34. The impact of lactate on the metabolic phenotype of tumor-associated macrophages
- Author
-
Xenia Geeraerts, Yvon Elkrim, Dorien Broekaert, Sarah-Maria Fendt, and Jo Van Ginderachter
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.