Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Trends Cancer ; 10(3): 175-176, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38355357

ABSTRACT

In a recent study, Kerzel et al. report a novel therapeutic strategy to engineer tumor-associated macrophages (TAMs) in vivo by inducing the expression of IFNα in these cells. This approach enables improved antigen presentation and T cell activation, leading to controlled tumor growth in multiple murine models of liver metastasis.


Subject(s)
Liver Neoplasms , Macrophages , Humans , Animals , Mice , Macrophages/metabolism , Liver Neoplasms/pathology , Immunotherapy
2.
Cell Rep ; 39(8): 110865, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35613577

ABSTRACT

Tissue-resident macrophages adapt to local signals within tissues to acquire specific functions. Neoplasia transforms the tissue, raising the question as to how the environmental perturbations contribute to tumor-associated macrophage (TAM) identity and functions. Combining single-cell RNA sequencing (scRNA-seq) with spatial localization of distinct TAM subsets by imaging, we discover that TAM transcriptomic programs follow two main differentiation paths according to their localization in the stroma or in the neoplastic epithelium of the mammary duct. Furthermore, this diversity is exclusively detected in a spontaneous tumor model and tracks the different tissue territories as well as the type of tumor lesion. These TAM subsets harbor distinct capacity to activate CD8+ T cells and phagocyte tumor cells, supporting that specific tumor regions, rather than defined activation states, are the major drivers of TAM plasticity and heterogeneity. The distinctions created here provide a framework to design cancer treatment targeting specific TAM niches.


Subject(s)
Breast Neoplasms , Tumor-Associated Macrophages , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , Macrophages/pathology , Transcriptome/genetics
3.
J Exp Med ; 217(11)2020 11 02.
Article in English | MEDLINE | ID: mdl-32648893

ABSTRACT

Wallerian degeneration (WD) is a process of autonomous distal degeneration of axons upon injury. Macrophages (MPs) of the peripheral nervous system (PNS) are the main cellular agent controlling this process. Some evidence suggests that resident PNS-MPs along with MPs of hematogenous origin may be involved, but whether these two subsets exert distinct functions is unknown. Combining MP-designed fluorescent reporter mice and coherent anti-Stokes Raman scattering (CARS) imaging of the sciatic nerve, we deciphered the spatiotemporal choreography of resident and recently recruited MPs after injury and unveiled distinct functions of these subsets, with recruited MPs being responsible for efficient myelin stripping and clearance and resident MPs being involved in axonal regrowth. This work provides clues to tackle selectively cellular processes involved in neurodegenerative diseases.


Subject(s)
Macrophages/immunology , Wallerian Degeneration/diagnostic imaging , Wallerian Degeneration/immunology , Animals , Axons/physiology , Disease Models, Animal , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/physiology , Nonlinear Optical Microscopy , Remyelination/genetics , Sciatic Nerve/diagnostic imaging , Sciatic Nerve/immunology , Sciatic Nerve/injuries , Transcriptome
4.
Front Immunol ; 11: 675, 2020.
Article in English | MEDLINE | ID: mdl-32425929

ABSTRACT

Sepsis is characterized by a systemic inflammation that can cause an immune dysfunction, for which the underlying mechanisms are unclear. We investigated the impact of cecal ligature and puncture (CLP)-mediated polymicrobial sepsis on monocyte (Mo) mobilization and functions. Our results show that CLP led to two consecutive phases of Mo deployment. The first one occurred within the first 3 days after the induction of the peritonitis, while the second phase was of a larger amplitude and extended up to a month after apparent clinical recovery. The latter was associated with the expansion of Mo in the tissue reservoirs (bone marrow and spleen), their release in the blood and their accumulation in the vasculature of peripheral non-lymphoid tissues. It occurred even after antibiotic treatment but relied on inflammatory-dependent pathways and inversely correlated with increased susceptibility and severity to a secondary infection. The intravascular lung Mo displayed limited activation capacity, impaired phagocytic functions and failed to transfer efficient protection against a secondary infection into monocytopenic CCR2-deficient mice. In conclusion, our work unveiled key dysfunctions of intravascular inflammatory Mo during the recovery phase of sepsis and provided new insights to improve patient protection against secondary infections.


Subject(s)
Inflammation/immunology , Monocytes/immunology , Sepsis/immunology , Animals , Antigens, Ly/analysis , CX3C Chemokine Receptor 1/physiology , Lung/immunology , Mice , Mice, Inbred C57BL , Monocytes/physiology , Neutrophils/immunology , Phagocytosis , Receptors, CCR2/physiology
5.
Front Immunol ; 10: 1799, 2019.
Article in English | MEDLINE | ID: mdl-31417566

ABSTRACT

Tumor-associated macrophages (TAM) represent the main immune cell population of the tumor microenvironment in most cancer. For decades, TAM have been the focus of intense investigation to understand how they modulate the tumor microenvironment and their implication in therapy failure. One consensus is that TAM are considered to exclusively originate from circulating monocyte precursors released from the bone marrow, fitting the original dogma of tissue-resident macrophage ontogeny. A second consensus proposed that TAM harbor either a classically activated M1 or alternatively activated M2 polarization profile, with almost opposite anti- and pro-tumoral activity respectively. These fundamental pillars are now revised in face of the latest discoveries on macrophage biology. Embryonic-derived macrophages were recently characterized as major contributors to the pool of tissue-resident macrophages in many tissues. Their turnover with macrophages derived from precursors of adult hematopoiesis seems to follow a regulation at the subtissular level. This has shed light on an ever more complex macrophage diversity in the tumor microenvironment than once thought and raise the question of their respective implication in tumor development compared to classical monocyte-derived macrophages. These recent advances highlight that TAM have actually not fully revealed their usefulness and deserve to be reconsidered. Understanding the link between TAM ontogeny and their various functions in tumor growth and interaction with the immune system represents one of the future challenges for cancer therapy.


Subject(s)
Macrophages/immunology , Monocytes/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Humans , Macrophages/pathology , Monocytes/pathology , Neoplasms/pathology , Neoplasms/therapy
6.
Front Immunol ; 10: 1201, 2019.
Article in English | MEDLINE | ID: mdl-31214174

ABSTRACT

In most cancers, myeloid cells represent the major component of the immune microenvironment. Deciphering the impact of these cells on tumor growth and in response to various anti-tumor therapies is a key issue. Many studies have elucidated the role of tumor-associated monocytes and tumor-associated macrophages (TAM) in tumor development, angiogenesis, and therapeutic failure. In contrast, tumor dendritic cells (DC) are associated with tumor antigen uptake and T-cell priming. Myeloid subpopulations display differences in ontogeny, state of differentiation and distribution within the neoplastic tissue, making them difficult to study. The development of high-dimensional genomic and cytometric analyses has unveiled the large functional diversity of myeloid cells. Important fundamental insights on the biology of myeloid cells have also been provided by a boom in functional fluorescent imaging techniques, in particular for TAM. These approaches allow the tracking of cell behavior in native physiological environments, incorporating spatio-temporal dimensions in the study of their functional activity. Nevertheless, tracking myeloid cells within the TME remains a challenging process as many markers overlap between monocytes, macrophages, DC, and neutrophils. Therefore, perfect discrimination between myeloid subsets remains impossible to date. Herein we review the specific functions of myeloid cells in tumor development unveiled by image-based tracking, the limits of fluorescent reporters commonly used to accurately track specific myeloid cells, and novel combinations of myeloid-associated fluorescent reporters that better discriminate the relative contributions of these cells to tumor biology according to their origin and tissue localization.


Subject(s)
Macrophages/immunology , Microscopy, Fluorescence/methods , Monocytes/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Animals , Cell Differentiation , Humans
7.
Cancer Immunol Res ; 7(3): 376-387, 2019 03.
Article in English | MEDLINE | ID: mdl-30696630

ABSTRACT

Radiotherapy (RT) represents one of the main anticancer approaches for the treatment of solid tumors. Beyond the expected direct effects of RT on tumor cells, evidence supporting the importance of an immune response to RT is growing. The balance between RT-mediated immunogenic and tolerogenic activity is ill-defined and deserves more attention. Herein, a murine model of head and neck squamous cell carcinoma was used to demonstrate that RT upregulated CCL2 chemokine production in tumor cells, leading to a CCR2-dependent accumulation of tumor necrosis factor alpha (TNFα)-producing monocytes and CCR2+ regulatory T cells (Treg). This corecruitment was associated with a TNFα-dependent activation of Tregs, dampening the efficacy of RT. Our results highlight an unexpected cross-talk between innate and adaptive immune system components and indicate CCL2/CCR2 and TNFα as potential clinical candidates to counterbalance the radioprotective action of monocyte-derived cells and Tregs, paving the way for potent combined radioimmunotherapies.


Subject(s)
Monocytes/immunology , Radiation Tolerance/immunology , Receptors, CCR2/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Disease Models, Animal , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/radiotherapy , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/radiotherapy , T-Lymphocytes, Regulatory/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
8.
J Cell Biol ; 217(8): 2931-2950, 2018 08 06.
Article in English | MEDLINE | ID: mdl-29941474

ABSTRACT

Understanding the mechanisms of cellular differentiation is challenging because differentiation is initiated by signaling pathways that drive temporally dynamic processes, which are difficult to analyze in vivo. We establish a new tool, Timer of cell kinetics and activity (Tocky; or toki [time in Japanese]). Tocky uses the fluorescent Timer protein, which spontaneously shifts its emission spectrum from blue to red, in combination with computer algorithms to reveal the dynamics of differentiation in vivo. Using a transcriptional target of T cell receptor (TCR) signaling, we establish Nr4a3-Tocky to follow downstream effects of TCR signaling. Nr4a3-Tocky reveals the temporal sequence of events during regulatory T cell (Treg) differentiation and shows that persistent TCR signals occur during Treg generation. Remarkably, antigen-specific T cells at the site of autoimmune inflammation also show persistent TCR signaling. In addition, by generating Foxp3-Tocky, we reveal the in vivo dynamics of demethylation of the Foxp3 gene. Thus, Tocky is a tool for cell biologists to address previously inaccessible questions by directly revealing dynamic processes in vivo.


Subject(s)
Cell Differentiation/genetics , Forkhead Transcription Factors/genetics , Gene Expression Profiling/methods , Algorithms , Animals , Demethylation , Forkhead Transcription Factors/metabolism , Kinetics , Luminescent Proteins/analysis , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Time Factors
9.
J Exp Med ; 215(10): 2536-2553, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30201786

ABSTRACT

Tissue-resident macrophages can self-maintain without contribution of adult hematopoiesis. Herein we show that tissue-resident interstitial macrophages (Res-TAMs) in mouse lungs contribute to the pool of tumor-associated macrophages (TAMs) together with CCR2-dependent recruited macrophages (MoD-TAMs). Res-TAMs largely correlated with tumor cell growth in vivo, while MoD-TAMs accumulation was associated with enhanced tumor spreading. Both cell subsets were depleted after chemotherapy, but MoD-TAMs rapidly recovered and performed phagocytosis-mediated tumor clearance. Interestingly, anti-VEGF treatment combined with chemotherapy inhibited both Res and Mod-TAM reconstitution without affecting monocyte infiltration and improved its efficacy. Our results reveal that the developmental origin of TAMs dictates their relative distribution, function, and response to cancer therapies in lung tumors.


Subject(s)
Lung Neoplasms/immunology , Macrophages/immunology , Monocytes/immunology , Phagocytosis , Animals , Lung Neoplasms/pathology , Macrophages/pathology , Mice , Mice, Knockout , Monocytes/pathology , Receptors, CCR2/immunology
SELECTION OF CITATIONS
SEARCH DETAIL