Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Cell ; 186(4): 764-785.e21, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36803604

ABSTRACT

The choroid plexus (ChP) is the blood-cerebrospinal fluid (CSF) barrier and the primary source of CSF. Acquired hydrocephalus, caused by brain infection or hemorrhage, lacks drug treatments due to obscure pathobiology. Our integrated, multi-omic investigation of post-infectious hydrocephalus (PIH) and post-hemorrhagic hydrocephalus (PHH) models revealed that lipopolysaccharide and blood breakdown products trigger highly similar TLR4-dependent immune responses at the ChP-CSF interface. The resulting CSF "cytokine storm", elicited from peripherally derived and border-associated ChP macrophages, causes increased CSF production from ChP epithelial cells via phospho-activation of the TNF-receptor-associated kinase SPAK, which serves as a regulatory scaffold of a multi-ion transporter protein complex. Genetic or pharmacological immunomodulation prevents PIH and PHH by antagonizing SPAK-dependent CSF hypersecretion. These results reveal the ChP as a dynamic, cellularly heterogeneous tissue with highly regulated immune-secretory capacity, expand our understanding of ChP immune-epithelial cell cross talk, and reframe PIH and PHH as related neuroimmune disorders vulnerable to small molecule pharmacotherapy.


Subject(s)
Choroid Plexus , Hydrocephalus , Humans , Blood-Brain Barrier/metabolism , Brain/metabolism , Choroid Plexus/metabolism , Hydrocephalus/cerebrospinal fluid , Hydrocephalus/immunology , Immunity, Innate , Cytokine Release Syndrome/pathology
2.
Immunity ; 57(6): 1225-1242.e6, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38749446

ABSTRACT

Classical monocytes (CMs) are ephemeral myeloid immune cells that circulate in the blood. Emerging evidence suggests that CMs can have distinct ontogeny and originate from either granulocyte-monocyte- or monocyte-dendritic-cell progenitors (GMPs or MDPs). Here, we report surface markers that allowed segregation of murine GMP- and MDP-derived CMs, i.e., GMP-Mo and MDP-Mo, as well as their functional characterization, including fate definition following adoptive cell transfer. GMP-Mo and MDP-Mo yielded an equal increase in homeostatic CM progeny, such as blood-resident non-classical monocytes and gut macrophages; however, these cells differentially seeded various other selected tissues, including the dura mater and lung. Specifically, GMP-Mo and MDP-Mo differentiated into distinct interstitial lung macrophages, linking CM dichotomy to previously reported pulmonary macrophage heterogeneity. Collectively, we provide evidence for the existence of two functionally distinct CM subsets in the mouse that differentially contribute to peripheral tissue macrophage populations in homeostasis and following challenge.


Subject(s)
Cell Differentiation , Macrophages , Monocytes , Animals , Monocytes/immunology , Monocytes/cytology , Mice , Cell Differentiation/immunology , Macrophages/immunology , Macrophages/metabolism , Lung/cytology , Lung/immunology , Homeostasis , Mice, Inbred C57BL , Dendritic Cells/immunology , Cell Lineage , Adoptive Transfer
3.
Immunity ; 55(11): 2085-2102.e9, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36228615

ABSTRACT

Microglia and border-associated macrophages (BAMs) are brain-resident self-renewing cells. Here, we examined the fate of microglia, BAMs, and recruited macrophages upon neuroinflammation and through resolution. Upon infection, Trypanosoma brucei parasites invaded the brain via its border regions, triggering brain barrier disruption and monocyte infiltration. Fate mapping combined with single-cell sequencing revealed microglia accumulation around the ventricles and expansion of epiplexus cells. Depletion experiments using genetic targeting revealed that resident macrophages promoted initial parasite defense and subsequently facilitated monocyte infiltration across brain barriers. These recruited monocyte-derived macrophages outnumbered resident macrophages and exhibited more transcriptional plasticity, adopting antimicrobial gene expression profiles. Recruited macrophages were rapidly removed upon disease resolution, leaving no engrafted monocyte-derived cells in the parenchyma, while resident macrophages progressively reverted toward a homeostatic state. Long-term transcriptional alterations were limited for microglia but more pronounced in BAMs. Thus, brain-resident and recruited macrophages exhibit diverging responses and dynamics during infection and resolution.


Subject(s)
Macrophages , Neuroinflammatory Diseases , Humans , Macrophages/metabolism , Monocytes/metabolism , Microglia/metabolism , Brain
4.
Immunity ; 54(1): 176-190.e7, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33333014

ABSTRACT

The developmental and molecular heterogeneity of tissue macrophages is unravelling, as are their diverse contributions to physiology and pathophysiology. Moreover, also given tissues harbor macrophages in discrete anatomic locations. Functional contributions of specific cell populations can in mice be dissected using Cre recombinase-mediated mutagenesis. However, single promoter-based Cre models show limited specificity for cell types. Focusing on macrophages in the brain, we establish here a binary transgenic system involving complementation-competent NCre and CCre fragments whose expression is driven by distinct promoters: Sall1ncre: Cx3cr1ccre mice specifically target parenchymal microglia and compound transgenic Lyve1ncre: Cx3cr1ccre animals target vasculature-associated macrophages, in the brain, as well as other tissues. We imaged the respective cell populations and retrieved their specific translatomes using the RiboTag in order to define them and analyze their differential responses to a challenge. Collectively, we establish the value of binary transgenesis to dissect tissue macrophage compartments and their functions.


Subject(s)
Brain/cytology , Central Nervous System/physiology , Integrases/metabolism , Macrophages/physiology , Microglia/physiology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity
5.
Immunity ; 53(5): 1033-1049.e7, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33049219

ABSTRACT

Microglia, the resident macrophages of the brain parenchyma, are key players in central nervous system (CNS) development, homeostasis, and disorders. Distinct brain pathologies seem associated with discrete microglia activation modules. How microglia regain quiescence following challenges remains less understood. Here, we explored the role of the interleukin-10 (IL-10) axis in restoring murine microglia homeostasis following a peripheral endotoxin challenge. Specifically, we show that lipopolysaccharide (LPS)-challenged mice harboring IL-10 receptor-deficient microglia displayed neuronal impairment and succumbed to fatal sickness. Addition of a microglial tumor necrosis factor (TNF) deficiency rescued these animals, suggesting a microglia-based circuit driving pathology. Single cell transcriptome analysis revealed various IL-10 producing immune cells in the CNS, including most prominently Ly49D+ NK cells and neutrophils, but not microglia. Collectively, we define kinetics of the microglia response to peripheral endotoxin challenge, including their activation and robust silencing, and highlight the critical role of non-microglial IL-10 in preventing deleterious microglia hyperactivation.


Subject(s)
Endotoxins/immunology , Interleukin-10/metabolism , Microglia/immunology , Microglia/metabolism , Animals , Biomarkers , Brain/immunology , Brain/metabolism , Brain/pathology , Cells, Cultured , Immunophenotyping , Interleukin-10/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lipopolysaccharides/immunology , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Mice
6.
Eur J Immunol ; 50(3): 459-463, 2020 03.
Article in English | MEDLINE | ID: mdl-31785096

ABSTRACT

Ligand-dependent Cre recombinases such as the CreERT2 system allow for tamoxifen-inducible Cre recombination. Important examples are the Cx3cr1-CreERT2 and Sall1-CreERT2 lines that are widely used for fate mapping and gene deletion studies of brain macrophages. Our results now show that both CreERT2 lines can exhibit a high rate of tamoxifen-independent "leaky" excision with some reporter strains, while this is not observed with others. We suggest that this disparity is determined by the length of the floxed transcriptional STOP cassette that is incorporated in the various reporter lines. In addition, the rate of spontaneous recombination was also determined by the CreERT2 expression levels and the longevity of the CreERT2-expressing cells. The implications of these results are discussed in the context of fate mapping and inducible gene deletion studies in macrophages and microglia.


Subject(s)
Integrases , Mice, Transgenic , Microglia , Models, Animal , Recombination, Genetic , Animals , Gene Deletion , Mice , Tamoxifen
7.
Cell Immunol ; 362: 104296, 2021 04.
Article in English | MEDLINE | ID: mdl-33556903

ABSTRACT

Efficient priming of anti-tumor T cells requires the uptake and presentation of tumor antigens by immunogenic dendritic cells (DCs) and occurs mainly in lymph nodes draining the tumor (tdLNs). However, tumors expand and activate myeloid-derived suppressor cells (MDSCs) that inhibit CTL functions by several mechanisms. While the immune-suppressive nature of the tumor microenvironment is largely documented, it is not known whether similar immune-suppressive mechanisms operate in the tdLNs. In this study, we analyzed MDSC characteristics within tdLNs. We show that, in a metastasis-free context, MO-MDSCs are the dominant MDSC population within tdLNs, that they are highly suppressive and that tumor proximity enhances their recruitment to tdLN via a CCR2/CCL2-dependent pathway. Altogether our results uncover a mechanism by which tumors evade the immune system that involves MDSC-mediated recruitment to the tdLN and the inhibition of T-cell activation even before reaching the highly immunosuppressive tumor microenvironment.


Subject(s)
Myeloid-Derived Suppressor Cells/metabolism , Receptors, CCR2/metabolism , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Female , Humans , Lymph Nodes/metabolism , Lymph Nodes/physiology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Myeloid Cells/immunology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/physiology , Neoplasms/immunology , Receptors, CCR2/immunology
8.
Glia ; 68(12): 2643-2660, 2020 12.
Article in English | MEDLINE | ID: mdl-32645232

ABSTRACT

Increasing evidence suggests that functional impairments at the level of the neurovascular unit (NVU) underlie many neurodegenerative and neuroinflammatory diseases. While being part of the NVU, astrocytes have been largely overlooked in this context and only recently, tightening of the glia limitans has been put forward as an important neuroprotective response to limit these injurious processes. In this study, using the retina as a central nervous system (CNS) model organ, we investigated the structure and function of the glia limitans, and reveal that the blood-retina barrier and glia limitans function as a coordinated double barrier to limit infiltration of leukocytes and immune molecules. We provide in vitro and in vivo evidence for a protective response at the NVU upon CNS injury, which evokes inflammation-induced glia limitans tightening. Matrix metalloproteinase-3 (MMP-3) was found to be a crucial regulator of this process, thereby revealing its beneficial and immunomodulatory role in the CNS. in vivo experiments in which MMP-3 activity was deleted via genetic and pharmacological approaches, combined with a comprehensive study of tight junction molecules, glial end feet markers, myeloid cell infiltration, cytokine expression and neurodegeneration, show that MMP-3 attenuates neuroinflammation and neurodegeneration by tightening the glia limitans, thereby pointing to a prominent role of MMP-3 in preserving the integrity of the NVU upon injury. Finally, we gathered promising evidence to suggest that IL1b, which is also regulated by MMP-3, is at least one of the molecular messengers that induces glia limitans tightening in the injured CNS.


Subject(s)
Optic Nerve Injuries , Astrocytes , Humans , Matrix Metalloproteinase 3 , Neuroglia , Retina
9.
Circulation ; 138(5): 494-508, 2018 07 31.
Article in English | MEDLINE | ID: mdl-29626067

ABSTRACT

BACKGROUND: Peripheral vascular resistance has a major impact on arterial blood pressure levels. Endothelial C-type natriuretic peptide (CNP) participates in the local regulation of vascular tone, but the target cells remain controversial. The cGMP-producing guanylyl cyclase-B (GC-B) receptor for CNP is expressed in vascular smooth muscle cells (SMCs). However, whereas endothelial cell-specific CNP knockout mice are hypertensive, mice with deletion of GC-B in vascular SMCs have unaltered blood pressure. METHODS: We analyzed whether the vasodilating response to CNP changes along the vascular tree, ie, whether the GC-B receptor is expressed in microvascular types of cells. Mice with a floxed GC-B ( Npr2) gene were interbred with Tie2-Cre or PDGF-Rß-Cre ERT2 lines to develop mice lacking GC-B in endothelial cells or in precapillary arteriolar SMCs and capillary pericytes. Intravital microscopy, invasive and noninvasive hemodynamics, fluorescence energy transfer studies of pericyte cAMP levels in situ, and renal physiology were combined to dissect whether and how CNP/GC-B/cGMP signaling modulates microcirculatory tone and blood pressure. RESULTS: Intravital microscopy studies revealed that the vasodilatatory effect of CNP increases toward small-diameter arterioles and capillaries. CNP consistently did not prevent endothelin-1-induced acute constrictions of proximal arterioles, but fully reversed endothelin effects in precapillary arterioles and capillaries. Here, the GC-B receptor is expressed both in endothelial and mural cells, ie, in pericytes. It is notable that the vasodilatatory effects of CNP were preserved in mice with endothelial GC-B deletion, but abolished in mice lacking GC-B in microcirculatory SMCs and pericytes. CNP, via GC-B/cGMP signaling, modulates 2 signaling cascades in pericytes: it activates cGMP-dependent protein kinase I to phosphorylate downstream targets such as the cytoskeleton-associated vasodilator-activated phosphoprotein, and it inhibits phosphodiesterase 3A, thereby enhancing pericyte cAMP levels. These pathways ultimately prevent endothelin-induced increases of pericyte calcium levels and pericyte contraction. Mice with deletion of GC-B in microcirculatory SMCs and pericytes have elevated peripheral resistance and chronic arterial hypertension without a change in renal function. CONCLUSIONS: Our studies indicate that endothelial CNP regulates distal arteriolar and capillary blood flow. CNP-induced GC-B/cGMP signaling in microvascular SMCs and pericytes is essential for the maintenance of normal microvascular resistance and blood pressure.


Subject(s)
Arterial Pressure/drug effects , Endothelial Cells/drug effects , Hypertension/metabolism , Microcirculation/drug effects , Microvessels/drug effects , Natriuretic Peptide, C-Type/pharmacology , Pericytes/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Biosensing Techniques , Calcium Signaling/drug effects , Cells, Cultured , Cyclic GMP/metabolism , Endothelial Cells/metabolism , Fluorescence Resonance Energy Transfer , Genetic Predisposition to Disease , Hypertension/genetics , Hypertension/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Microvessels/physiopathology , Natriuretic Peptide, C-Type/metabolism , Paracrine Communication/drug effects , Phenotype , Receptor, Platelet-Derived Growth Factor beta/deficiency , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/genetics
10.
Cell Immunol ; 330: 188-201, 2018 08.
Article in English | MEDLINE | ID: mdl-29482836

ABSTRACT

Tumors of various histological origins show abundant infiltration of myeloid cells from early stages of disease progression. These cells have a profound impact on antitumor immunity and influence fundamental processes that underlie malignancy, including neoangiogenesis, sustained cancer cell proliferation, metastasis and therapy resistance. For these reasons, development of therapeutic approaches to deplete or reprogram myeloid cells in cancer is an emerging field of interest. However, knowledge about the heterogeneity of myeloid cells in tumors and their variability between patients and disease stages is still limited. In this review, we summarize the most recent advances in our understanding about how the phenotype of tumor-associated macrophages, monocytes, neutrophils, myeloid-derived suppressor cells and dendritic cells is dictated by their ontogeny, activation status and localization. We also outline major open questions that will only be resolved by applying high-dimensional single-cell technologies and systems biology approaches in the analysis of the tumor microenvironment.


Subject(s)
Cell Proliferation , Myeloid Cells/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Dendritic Cells/immunology , Humans , Macrophages/immunology , Monocytes/immunology , Myeloid Cells/pathology , Neoplasms/pathology , Neutrophils/immunology
11.
Bioconjug Chem ; 29(7): 2394-2405, 2018 07 18.
Article in English | MEDLINE | ID: mdl-29889515

ABSTRACT

Tumor-associated macrophages (TAMs) with high expression levels of the Macrophage Mannose Receptor (MMR, CD206) exhibit a strong angiogenic and immune suppressive activity. Thus, they are a highly attractive target in cancer immunotherapy, with the aim to modulate their protumoral behavior. Here, we introduce polymer nanogels as potential drug nanocarriers which were site-specifically decorated with a Nanobody (Nb) specific for the MMR. Using azide-functionalized RAFT chain transfer agents, they provide access to amphiphilic reactive ester block copolymers that self-assemble into micelles and are afterwards core-cross-linked toward fully hydrophilic nanogels with terminal azide groups on their surface. MMR-targeting Nb can site-selectively be functionalized with one single cyclooctyne moiety by maleimide-cysteine chemistry under mildly reducing conditions which enables successful chemoorthogonal conjugation to the nanogels. The resulting Nb-functionalized nanogels were highly efficient in targeting MMR-expressing cells and TAMs both in vitro and in vivo. We believe that these findings pave the road for targeted eradication or modulation of pro-tumoral MMRhigh TAMs.


Subject(s)
Antibodies, Monoclonal/immunology , Drug Carriers/chemical synthesis , Immunotherapy/methods , Lectins, C-Type/immunology , Macrophages/drug effects , Mannose-Binding Lectins/immunology , Neoplasms/therapy , Receptors, Cell Surface/immunology , Alkynes , Azides , Cycloaddition Reaction , Humans , Mannose Receptor , Micelles , Neoplasms/immunology , Polymers
12.
Biotechnol Bioeng ; 115(7): 1778-1792, 2018 07.
Article in English | MEDLINE | ID: mdl-29573361

ABSTRACT

Functional mosaic analysis allows for the direct comparison of mutant cells with differentially marked control cells in the same organism. While this offers a powerful approach for elucidating the role of specific genes or signalling pathways in cell populations of interest, genetic strategies for generating functional mosaicism remain challenging. We describe a novel and streamlined approach for functional mosaic analysis, which combines stochastic Cre/lox recombination with gene targeting in the ROSA26 locus. With the RoMo strategy a cell population of interest is randomly split into a cyan fluorescent and red fluorescent subset, of which the latter overexpresses a chosen transgene. To integrate this approach into high-throughput gene targeting initiatives, we developed a procedure that utilizes Gateway cloning for the generation of new targeting vectors. RoMo can be used for gain-of-function experiments or for altering signaling pathways in a mosaic fashion. To demonstrate this, we developed RoMo-dnGs mice, in which Cre-recombined red fluorescent cells co-express a dominant-negative Gs protein. RoMo-dnGs mice allowed us to inhibit G protein-coupled receptor activation in a fraction of cells, which could then be directly compared to differentially marked control cells in the same animal. We demonstrate how RoMo-dnGs mice can be used to obtain mosaicism in the brain and in peripheral organs for various cell types. RoMo offers an efficient new approach for functional mosaic analysis that extends the current toolbox and may reveal important new insights into in vivo gene function.


Subject(s)
Gene Targeting/methods , Genetic Loci , RNA, Untranslated/genetics , Recombination, Genetic , Animals , Integrases/metabolism , Mice , Mosaicism
13.
Biochim Biophys Acta ; 1865(1): 23-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26145884

ABSTRACT

The tumor-promoting role of macrophages has been firmly established in most cancer types. However, macrophage identity has been a matter of debate, since several levels of complexity result in considerable macrophage heterogeneity. Ontogenically, tissue-resident macrophages derive from yolk sac progenitors which either directly or via a fetal liver monocyte intermediate differentiate into distinct macrophage types during embryogenesis and are maintained throughout life, while a disruption of the steady state mobilizes monocytes and instructs the formation of monocyte-derived macrophages. Histologically, the macrophage phenotype is heavily influenced by the tissue microenvironment resulting in molecularly and functionally distinct macrophages in distinct organs. Finally, a change in the tissue microenvironment as a result of infectious or sterile inflammation instructs different modes of macrophage activation. These considerations are relevant in the context of tumors, which can be considered as sites of chronic sterile inflammation encompassing subregions with distinct environmental conditions (for example, hypoxic versus normoxic). Here, we discuss existing evidence on the role of macrophage subpopulations in steady state tissue and primary tumors of the breast, lung, pancreas, brain and liver.


Subject(s)
Macrophages/physiology , Monocytes/cytology , Neoplasms/immunology , Tumor Microenvironment , Animals , Humans
14.
Curr Opin Oncol ; 29(1): 55-61, 2017 01.
Article in English | MEDLINE | ID: mdl-27792052

ABSTRACT

PURPOSE OF REVIEW: Tumors contain not only cancer cells but also nontransformed types of cells, the stromal cells. A bidirectional interplay exists between transformed and nontransformed cells leading to tumor progression and metastasis. Tumor-associated macrophages (TAMs) are the most abundant tumor-infiltrating leukocytes characterized by a high heterogeneity and plasticity. TAMs exhibit strong protumoral activities and are related to bad prognosis and worse overall survival in various cancer types. RECENT FINDINGS: Recent progress has delineated the existence of distinct TAM subsets in primary tumors and metastatic sites regulated by diverse mechanisms and triggering strong protumoral functions such as immunossuppression, angiogenesis, metastasis and resistance to current therapies. SUMMARY: Delineating the regulatory pathways governing TAM heterogeneity and activation could present a novel frontier in cancer therapy. TAM targeting/repolarization is considered as a promising novel therapeutic modality in combination with standard-of-care therapies or immuno checkpoint blockers.


Subject(s)
Macrophages/pathology , Neoplasms/pathology , Tumor Microenvironment , Animals , Cell Transformation, Neoplastic , Disease Progression , Humans
15.
Eur J Immunol ; 43(11): 2930-42, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23878002

ABSTRACT

Tumor growth coincides with an accumulation of myeloid-derived suppressor cells (MDSCs), which exert immune suppression and which consist of two main subpopulations, known as monocytic (MO) CD11b(+) CD115(+) Ly6G(-) Ly6C(high) MDSCs and granulocytic CD11b(+) CD115(-) Ly6G(+) Ly6C(int) polymorphonuclear (PMN)-MDSCs. However, whether these distinct MDSC subsets hamper all aspects of early CD8(+) T-cell activation--including cytokine production, surface marker expression, survival, and cytotoxicity--is currently unclear. Here, employing an in vitro coculture system, we demonstrate that splenic MDSC subsets suppress antigen-driven CD8(+) T-cell proliferation, but differ in their dependency on IFN-γ, STAT-1, IRF-1, and NO to do so. Moreover, MO-MDSC and PMN-MDSCs diminish IL-2 levels, but only MO-MDSCs affect IL-2Rα (CD25) expression and STAT-5 signaling. Unexpectedly, however, both MDSC populations stimulate IFN-γ production by CD8(+) T cells on a per cell basis, illustrating that some T-cell activation characteristics are actually stimulated by MDSCs. Conversely, MO-MDSCs counteract the activation-induced change in CD44, CD62L, CD162, and granzyme B expression, while promoting CD69 and Fas upregulation. Together, these effects result in an altered CD8(+) T-cell adhesiveness to the extracellular matrix and selectins, sensitivity to FasL-mediated apoptosis, and cytotoxicity. Hence, MDSCs intricately influence different CD8(+) T-cell activation events in vitro, whereby some parameters are suppressed while others are stimulated.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Ly/metabolism , Apoptosis/immunology , CD11b Antigen/metabolism , Cell Adhesion/immunology , Cell Line , Cell Proliferation , Female , Granzymes/biosynthesis , Hyaluronan Receptors/biosynthesis , Interferon Regulatory Factor-1 , Interferon-gamma/biosynthesis , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , L-Selectin/biosynthesis , Lectins, C-Type/biosynthesis , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Nitric Oxide/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , STAT1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/immunology , Up-Regulation , fas Receptor/biosynthesis
16.
Front Immunol ; 15: 1323409, 2024.
Article in English | MEDLINE | ID: mdl-38352874

ABSTRACT

Background: Alzheimer's disease (AD) is the most common neurodegenerative disorder affecting memory and cognition. The disease is accompanied by an abnormal deposition of ß-amyloid plaques in the brain that contributes to neurodegeneration and is known to induce glial inflammation. Studies in the APP/PS1 mouse model of ß-amyloid-induced neuropathology have suggested a role for inflammasome activation in ß-amyloid-induced neuroinflammation and neuropathology. Methods: Here, we evaluated the in vivo role of microglia-selective and full body inflammasome signalling in several mouse models of ß-amyloid-induced AD neuropathology. Results: Microglia-specific deletion of the inflammasome regulator A20 and inflammasome effector protease caspase-1 in the AppNL-G-F and APP/PS1 models failed to identify a prominent role for microglial inflammasome signalling in ß-amyloid-induced neuropathology. Moreover, global inflammasome inactivation through respectively full body deletion of caspases 1 and 11 in AppNL-G-F mice and Nlrp3 deletion in APP/PS1 mice also failed to modulate amyloid pathology and disease progression. In agreement, single-cell RNA sequencing did not reveal an important role for Nlrp3 signalling in driving microglial activation and the transition into disease-associated states, both during homeostasis and upon amyloid pathology. Conclusion: Collectively, these results question a generalizable role for inflammasome activation in preclinical amyloid-only models of neuroinflammation.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/pathology , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Neuroinflammatory Diseases , Mice, Transgenic , Amyloid , Amyloidogenic Proteins
17.
Front Immunol ; 14: 1183641, 2023.
Article in English | MEDLINE | ID: mdl-37275902

ABSTRACT

Despite relentless efforts to improve outcome, the prognosis of glioblastoma (GBM) remains poor. Standard therapy at first diagnosis consists of maximal safe surgical resection followed by radiochemotherapy, but treatment options at recurrence are scarce and have limited efficacy. Immunotherapy is a broad term that covers several treatment strategies, including immune checkpoint inhibition (ICI). The successes of systemically administered therapeutic monoclonal antibodies that block the Programmed death receptor or ligand (PD-(L)1) and Cytotoxic T-Lymphocyte associated protein (CTLA)-4 immune checkpoints in other cancer types could not be reproduced in glioblastoma. This is considered to be related to the intrinsic low immunogenicity and strong immunosuppressive tumor microenvironment of glioblastoma, in addition to the presence of a blood-glioma and blood-brain barrier that limits many systemically administered therapeutic agents from reaching their target. In this mini-review, we address the specific aspects of immune suppression in glioblastoma and discuss potential strategies that could help to overcome it. The potential advantages of incorporating surgical resection in clinical trials of immunotherapy for glioblastoma, including window-of-opportunity studies, are highlighted. Combination strategies that include surgical resection, as well as local administration of therapeutic agents in the brain are discussed as a potential strategy to achieve an effective immunological response against glioblastoma.


Subject(s)
Glioblastoma , Glioma , Humans , Glioblastoma/metabolism , Immunotherapy , Immunosuppression Therapy , Prognosis , Tumor Microenvironment
18.
Front Immunol ; 14: 1238452, 2023.
Article in English | MEDLINE | ID: mdl-37691953

ABSTRACT

The liver is a vital metabolic organ that also performs important immune-regulatory functions. In the context of infections, the liver represents a target site for various pathogens, while also having an outstanding capacity to filter the blood from pathogens and to contain infections. Pathogen scavenging by the liver is primarily performed by its large and heterogeneous macrophage population. The major liver-resident macrophage population is located within the hepatic microcirculation and is known as Kupffer cells (KCs). Although other minor macrophages reside in the liver as well, KCs remain the best characterized and are the best well-known hepatic macrophage population to be functionally involved in the clearance of infections. The response of KCs to pathogenic insults often governs the overall severity and outcome of infections on the host. Moreover, infections also impart long-lasting, and rarely studied changes to the KC pool. In this review, we discuss current knowledge on the biology and the various roles of liver macrophages during infections. In addition, we reflect on the potential of infection history to imprint long-lasting effects on macrophages, in particular liver macrophages.


Subject(s)
Communicable Diseases , Kupffer Cells , Humans , Liver , Macrophages , Kinetics
19.
Acta Neuropathol Commun ; 11(1): 85, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37226256

ABSTRACT

The multifaceted nature of neuroinflammation is highlighted by its ability to both aggravate and promote neuronal health. While in mammals retinal ganglion cells (RGCs) are unable to regenerate following injury, acute inflammation can induce axonal regrowth. However, the nature of the cells, cellular states and signalling pathways that drive this inflammation-induced regeneration have remained elusive. Here, we investigated the functional significance of macrophages during RGC de- and regeneration, by characterizing the inflammatory cascade evoked by optic nerve crush (ONC) injury, with or without local inflammatory stimulation in the vitreous. By combining single-cell RNA sequencing and fate mapping approaches, we elucidated the response of retinal microglia and recruited monocyte-derived macrophages (MDMs) to RGC injury. Importantly, inflammatory stimulation recruited large numbers of MDMs to the retina, which exhibited long-term engraftment and promoted axonal regrowth. Ligand-receptor analysis highlighted a subset of recruited macrophages that exhibited expression of pro-regenerative secreted factors, which were able to promote axon regrowth via paracrine signalling. Our work reveals how inflammation may promote CNS regeneration by modulating innate immune responses, providing a rationale for macrophage-centred strategies for driving neuronal repair following injury and disease.


Subject(s)
Axons , Optic Nerve Injuries , Animals , Retina , Retinal Ganglion Cells , Macrophages , Inflammation , Mammals
20.
Theranostics ; 13(1): 355-373, 2023.
Article in English | MEDLINE | ID: mdl-36593955

ABSTRACT

Rationale: Nanobodies (Nbs) have emerged as an elegant alternative to the use of conventional monoclonal antibodies in cancer therapy, but a detailed microscopic insight into the in vivo pharmacokinetics of different Nb formats in tumor-bearers is lacking. This is especially relevant for the recognition and targeting of pro-tumoral tumor-associated macrophages (TAMs), which may be located in less penetrable tumor regions. Methods: We employed anti-Macrophage Mannose Receptor (MMR) Nbs, in a monovalent (m) or bivalent (biv) format, to assess in vivo TAM targeting. Intravital and confocal microscopy were used to analyse the blood clearance rate and targeting kinetics of anti-MMR Nbs in tumor tissue, healthy muscle tissue and liver. Fluorescence Molecular Tomography was applied to confirm anti-MMR Nb accumulation in the primary tumor and in metastatic lesions. Results: Intravital microscopy demonstrated significant differences in the blood clearance rate and macrophage targeting kinetics of (m) and (biv)anti-MMR Nbs, both in tumoral and extra-tumoral tissue. Importantly, (m)anti-MMR Nbs are superior in reaching tissue macrophages, an advantage that is especially prominent in tumor tissue. The administration of a molar excess of unlabelled (biv)anti-MMR Nbs increased the (m)anti-MMR Nb bioavailability and impacted on its macrophage targeting kinetics, preventing their accumulation in extra-tumoral tissue (especially in the liver) but only partially influencing their interaction with TAMs. Finally, anti-MMR Nb administration not only allowed the visualization of TAMs in primary tumors, but also at a distant metastatic site. Conclusions: These data describe, for the first time, a microscopic analysis of (m) and (biv)anti-MMR Nb pharmacokinetics in tumor and healthy tissues. The concepts proposed in this study provide important knowledge for the future use of Nbs as diagnostic and therapeutic agents, especially for the targeting of tumor-infiltrating immune cells.


Subject(s)
Neoplasms , Single-Domain Antibodies , Humans , Mannose Receptor , Lectins, C-Type , Mannose-Binding Lectins , Receptors, Cell Surface , Tumor-Associated Macrophages , Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL