Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cell ; 175(2): 400-415.e13, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30173915

ABSTRACT

Macrophages are highly heterogeneous tissue-resident immune cells that perform a variety of tissue-supportive functions. The current paradigm dictates that intestinal macrophages are continuously replaced by incoming monocytes that acquire a pro-inflammatory or tissue-protective signature. Here, we identify a self-maintaining population of macrophages that arise from both embryonic precursors and adult bone marrow-derived monocytes and persists throughout adulthood. Gene expression and imaging studies of self-maintaining macrophages revealed distinct transcriptional profiles that reflect their unique localization (i.e., closely positioned to blood vessels, submucosal and myenteric plexus, Paneth cells, and Peyer's patches). Depletion of self-maintaining macrophages resulted in morphological abnormalities in the submucosal vasculature and loss of enteric neurons, leading to vascular leakage, impaired secretion, and reduced intestinal motility. These results provide critical insights in intestinal macrophage heterogeneity and demonstrate the strategic role of self-maintaining macrophages in gut homeostasis and intestinal physiology.


Subject(s)
Intestines/immunology , Macrophages/immunology , Animals , Body Patterning/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Gastrointestinal Motility/immunology , Gastrointestinal Motility/physiology , Homeostasis , Inflammation/immunology , Intestinal Mucosa/immunology , Intestine, Small/metabolism , Mice , Monocytes/metabolism , Neurons/metabolism , Phagocytes/immunology , Transcriptome
2.
Immunity ; 55(9): 1530-1548, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36103851

ABSTRACT

The gastrointestinal tract has the important task of absorbing nutrients, a complex process that requires an intact barrier allowing the passage of nutrients but that simultaneously protects the host against invading microorganisms. To maintain and regulate intestinal homeostasis, the gut is equipped with one of the largest populations of macrophages in the body. Here, we will discuss our current understanding of intestinal macrophage heterogeneity and describe their main functions in the different anatomical niches of the gut during steady state. In addition, their role in inflammatory conditions such as infection, inflammatory bowel disease, and postoperative ileus are discussed, highlighting the roles of macrophages in immune defense. To conclude, we describe the interaction between macrophages and the enteric nervous system during development and adulthood and highlight their contribution to neurodegeneration in the context of aging and diabetes.


Subject(s)
Enteric Nervous System , Inflammatory Bowel Diseases , Adult , Homeostasis , Humans , Macrophages
3.
Immunity ; 54(5): 850-852, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33979582

ABSTRACT

In this issue of Immunity, Schiller et al. report that local sympathetic nerve activation decreases endothelial expression of the adhesion molecule MAdCAM-1, reducing immune cell infiltration and colitis-induced inflammation. These findings suggest that local sympathetic stimulation provides a key gateway for regulating organ homeostasis.


Subject(s)
Colitis , Mucoproteins , Cell Adhesion Molecules , Endothelium , Humans , Neuroimmunomodulation
5.
Nature ; 618(7966): 818-826, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37316669

ABSTRACT

Correct development and maturation of the enteric nervous system (ENS) is critical for survival1. At birth, the ENS is immature and requires considerable refinement to exert its functions in adulthood2. Here we demonstrate that resident macrophages of the muscularis externa (MMϕ) refine the ENS early in life by pruning synapses and phagocytosing enteric neurons. Depletion of MMϕ before weaning disrupts this process and results in abnormal intestinal transit. After weaning, MMϕ continue to interact closely with the ENS and acquire a neurosupportive phenotype. The latter is instructed by transforming growth factor-ß produced by the ENS; depletion of the ENS and disruption of transforming growth factor-ß signalling result in a decrease in neuron-associated MMϕ associated with loss of enteric neurons and altered intestinal transit. These findings introduce a new reciprocal cell-cell communication responsible for maintenance of the ENS and indicate that the ENS, similarly to the brain, is shaped and maintained by a dedicated population of resident macrophages that adapts its phenotype and transcriptome to the timely needs of the ENS niche.


Subject(s)
Enteric Nervous System , Intestines , Macrophages , Enteric Nervous System/cytology , Enteric Nervous System/growth & development , Enteric Nervous System/physiology , Intestines/innervation , Lymphotoxin-alpha/metabolism , Macrophages/metabolism , Macrophages/physiology , Neurons/physiology , Weaning , Cell Communication , Transcriptome , Phenotype , Phagocytosis , Synapses , Neuronal Plasticity , Gastrointestinal Transit
6.
Nature ; 620(7974): 634-642, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37438525

ABSTRACT

The physiological functions of mast cells remain largely an enigma. In the context of barrier damage, mast cells are integrated in type 2 immunity and, together with immunoglobulin E (IgE), promote allergic diseases. Allergic symptoms may, however, facilitate expulsion of allergens, toxins and parasites and trigger future antigen avoidance1-3. Here, we show that antigen-specific avoidance behaviour in inbred mice4,5 is critically dependent on mast cells; hence, we identify the immunological sensor cell linking antigen recognition to avoidance behaviour. Avoidance prevented antigen-driven adaptive, innate and mucosal immune activation and inflammation in the stomach and small intestine. Avoidance was IgE dependent, promoted by Th2 cytokines in the immunization phase and by IgE in the execution phase. Mucosal mast cells lining the stomach and small intestine rapidly sensed antigen ingestion. We interrogated potential signalling routes between mast cells and the brain using mutant mice, pharmacological inhibition, neural activity recordings and vagotomy. Inhibition of leukotriene synthesis impaired avoidance, but overall no single pathway interruption completely abrogated avoidance, indicating complex regulation. Collectively, the stage for antigen avoidance is set when adaptive immunity equips mast cells with IgE as a telltale of past immune responses. On subsequent antigen ingestion, mast cells signal termination of antigen intake. Prevention of immunopathology-causing, continuous and futile responses against per se innocuous antigens or of repeated ingestion of toxins through mast-cell-mediated antigen-avoidance behaviour may be an important arm of immunity.


Subject(s)
Allergens , Avoidance Learning , Hypersensitivity , Mast Cells , Animals , Mice , Allergens/immunology , Avoidance Learning/physiology , Hypersensitivity/immunology , Immunoglobulin E/immunology , Mast Cells/immunology , Stomach/immunology , Vagotomy , Immunity, Innate/immunology , Immunity, Mucosal/immunology , Th2 Cells/immunology , Cytokines/immunology , Leukotrienes/biosynthesis , Leukotrienes/immunology , Intestine, Small/immunology
7.
Nature ; 590(7844): 151-156, 2021 02.
Article in English | MEDLINE | ID: mdl-33442055

ABSTRACT

Up to 20% of people worldwide develop gastrointestinal symptoms following a meal1, leading to decreased quality of life, substantial morbidity and high medical costs. Although the interest of both the scientific and lay communities in this issue has increased markedly in recent years, with the worldwide introduction of gluten-free and other diets, the underlying mechanisms of food-induced abdominal complaints remain largely unknown. Here we show that a bacterial infection and bacterial toxins can trigger an immune response that leads to the production of dietary-antigen-specific IgE antibodies in mice, which are limited to the intestine. Following subsequent oral ingestion of the respective dietary antigen, an IgE- and mast-cell-dependent mechanism induced increased visceral pain. This aberrant pain signalling resulted from histamine receptor H1-mediated sensitization of visceral afferents. Moreover, injection of food antigens (gluten, wheat, soy and milk) into the rectosigmoid mucosa of patients with irritable bowel syndrome induced local oedema and mast cell activation. Our results identify and characterize a peripheral mechanism that underlies food-induced abdominal pain, thereby creating new possibilities for the treatment of irritable bowel syndrome and related abdominal pain disorders.


Subject(s)
Abdominal Pain/immunology , Abdominal Pain/pathology , Allergens/immunology , Food Hypersensitivity/immunology , Food/adverse effects , Intestines/immunology , Irritable Bowel Syndrome/immunology , Abdominal Pain/etiology , Abdominal Pain/microbiology , Adult , Animals , Citrobacter rodentium/immunology , Diarrhea/immunology , Diarrhea/microbiology , Diarrhea/pathology , Enterobacteriaceae Infections/complications , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Female , Food Hypersensitivity/complications , Food Hypersensitivity/microbiology , Food Hypersensitivity/pathology , Glutens/immunology , Humans , Immunoglobulin E/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestines/microbiology , Intestines/pathology , Irritable Bowel Syndrome/etiology , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/pathology , Male , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Middle Aged , Milk/immunology , Ovalbumin/immunology , Quality of Life , Receptors, Histamine H1/metabolism , Soybean Proteins/immunology , Triticum/immunology
8.
Int Immunol ; 33(6): 337-348, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33788920

ABSTRACT

Accumulating evidence shows that intestinal homeostasis is mediated by cross-talk between the nervous system, enteric neurons and immune cells, together forming specialized neuroimmune units at distinct anatomical locations within the gut. In this review, we will particularly discuss how the intrinsic and extrinsic neuronal circuitry regulates macrophage function and phenotype in the gut during homeostasis and aberrant inflammation, such as observed in inflammatory bowel disease (IBD). Furthermore, we will provide an overview of basic and translational IBD research using these neuronal circuits as a novel therapeutic tool. Finally, we will highlight the different challenges ahead to make bioelectronic neuromodulation a standard treatment for intestinal immune-mediated diseases.


Subject(s)
Brain-Gut Axis/immunology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Animals , Homeostasis/immunology , Humans , Neurons/immunology
9.
Gut ; 68(8): 1406-1416, 2019 08.
Article in English | MEDLINE | ID: mdl-30472681

ABSTRACT

OBJECTIVES: Vagus nerve stimulation (VNS), most likely via enteric neurons, prevents postoperative ileus (POI) by reducing activation of alpha7 nicotinic receptor (α7nAChR) positive muscularis macrophages (mMφ) and dampening surgery-induced intestinal inflammation. Here, we evaluated if 5-HT4 receptor (5-HT4R) agonist prucalopride can mimic this effect in mice and human. DESIGN: Using Ca2+ imaging, the effect of electrical field stimulation (EFS) and prucalopride was evaluated in situ on mMφ activation evoked by ATP in jejunal muscularis tissue. Next, preoperative and postoperative administration of prucalopride (1-5 mg/kg) was compared with that of preoperative VNS in a model of POI in wild-type and α7nAChR knockout mice. Finally, in a pilot study, patients undergoing a Whipple procedure were preoperatively treated with prucalopride (n=10), abdominal VNS (n=10) or sham/placebo (n=10) to evaluate the effect on intestinal inflammation and clinical recovery of POI. RESULTS: EFS reduced the ATP-induced Ca2+ response of mMφ, an effect that was dampened by neurotoxins tetrodotoxin and ω-conotoxin and mimicked by prucalopride. In vivo, prucalopride administered before, but not after abdominal surgery reduced intestinal inflammation and prevented POI in wild-type, but not in α7nAChR knockout mice. In humans, preoperative administration of prucalopride, but not of VNS, decreased Il6 and Il8 expression in the muscularis externa and improved clinical recovery. CONCLUSION: Enteric neurons dampen mMφ activation, an effect mimicked by prucalopride. Preoperative, but not postoperative treatment with prucalopride prevents intestinal inflammation and shortens POI in both mice and human, indicating that preoperative administration of 5-HT4R agonists should be further evaluated as a treatment of POI. TRIAL REGISTRATION NUMBER: NCT02425774.


Subject(s)
Benzofurans , Ileus , Intestine, Small , Muscle, Smooth , Pancreaticoduodenectomy/adverse effects , Postoperative Complications , Adult , Animals , Benzofurans/administration & dosage , Benzofurans/pharmacology , Disease Models, Animal , Female , Gastrointestinal Motility/drug effects , Humans , Ileus/etiology , Ileus/immunology , Ileus/physiopathology , Ileus/prevention & control , Inflammation/immunology , Inflammation/prevention & control , Intestine, Small/immunology , Intestine, Small/innervation , Intestine, Small/pathology , Intestine, Small/physiopathology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Muscle, Smooth/drug effects , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Pancreaticoduodenectomy/methods , Pilot Projects , Postoperative Complications/immunology , Postoperative Complications/physiopathology , Postoperative Complications/prevention & control , Serotonin 5-HT4 Receptor Agonists/administration & dosage , Serotonin 5-HT4 Receptor Agonists/pharmacology , Treatment Outcome , alpha7 Nicotinic Acetylcholine Receptor/metabolism
10.
Allergy ; 74(9): 1748-1759, 2019 09.
Article in English | MEDLINE | ID: mdl-30897213

ABSTRACT

BACKGROUND: The vagus nerve has emerged as an important modulator of the intestinal immune system. Its anti-inflammatory properties have been previously shown in innate and Th1/Th17 predominant inflammatory models. To what extent the vagus nerve is of importance in Th2 inflammatory responses like food allergy is still unclear. In this study, we therefore aimed to investigate the effect of vagotomy (VGX) and vagus nerve stimulation (VNS), on the development and severity of experimental food allergy. METHODS: Balb/C mice were first sensitized with ovalbumin (OVA) in the presence of alum. Prior to oral challenges with OVA, mice were subjected to VGX or VNS. Disease severity was determined by assessing severity and onset of diarrhoea, OVA-specific antibody production, mast cell number and activity, inflammatory gene expression in duodenal tissue and lamina propria immune cells by flow cytometry analysis. RESULTS: When compared to control mice, VGX did not significantly affect the development and severity of the disease in our model of food allergy. VNS, on the other hand, resulted in a significant amelioration of the different inflammatory parameters assessed. This effect was independent of α7nAChR and is possibly mediated through the dampening of mast cells and increased phagocytosis of OVA by CX3CR1hi macrophages. CONCLUSIONS: These results underscore the anti-inflammatory properties of the vagus nerve and the potential of neuro-immune interactions in the intestine. Further insight into the underlying mechanisms could ultimately lead to novel therapeutic approaches in the treatment of not only food allergy but also other immune-mediated diseases.


Subject(s)
Food Hypersensitivity/etiology , Food Hypersensitivity/metabolism , Gastroenteritis/etiology , Gastroenteritis/metabolism , Vagus Nerve Stimulation , Allergens , Animals , Biomarkers , Cell Membrane Permeability , Disease Models, Animal , Food Hypersensitivity/diagnosis , Gastroenteritis/pathology , Immunophenotyping , Macrophages/immunology , Macrophages/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mastocytosis , Mice , Mice, Knockout , Neutrophil Infiltration/immunology , Ovalbumin/immunology , Severity of Illness Index , Vagotomy , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
11.
Cell Immunol ; 330: 142-150, 2018 08.
Article in English | MEDLINE | ID: mdl-29291892

ABSTRACT

Macrophages residing in the muscularis externa of the gastrointestinal tract are highly specialized cells that are essential for tissue homeostasis during steady-state conditions as well as during disease. They are characterized by their unique protective functional phenotype that is undoubtedly a consequence of the reciprocal interaction with their environment, including the enteric nervous system. This muscularis macrophage-neuron interaction dictates intestinal motility and promotes tissue-protection during injury and infection, but can also contribute to tissue damage in gastrointestinal disorders such as post-operative ileus and gastroparesis. Although the importance of muscularis macrophages is clearly recognized, different aspects of these cells remain largely unexplored such their origin, longevity and instructive signals that determine their function and phenotype. In this review, we will discuss the phenotype, functions and origin of muscularis macrophages during steady-state and disease conditions. We will highlight the bidirectional crosstalk with neurons and potential therapeutic strategies that target and manipulate muscularis macrophages to restore their protective signature as a treatment for disease.


Subject(s)
Homeostasis/immunology , Intestinal Diseases/immunology , Intestines/immunology , Macrophages/immunology , Muscle, Smooth/immunology , Animals , Enteric Nervous System/immunology , Gastrointestinal Motility/immunology , Humans , Intestines/innervation , Muscle, Smooth/cytology , Muscle, Smooth/innervation , Neurons/immunology
12.
Gut ; 66(12): 2098-2109, 2017 12.
Article in English | MEDLINE | ID: mdl-28615302

ABSTRACT

OBJECTIVE: Postoperative ileus (POI) is assumed to result from myeloid cells infiltrating the intestinal muscularis 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. DESIGN: IM 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. RESULTS: At early time points, IM-induced GI transit delay and inflammation were equal in WT and Ccr2 -/- mice. However, GI transit recovery after IM was significantly delayed in Ccr2 -/- 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 in Ccr2 -/- mice. CONCLUSION: Our 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.


Subject(s)
Ileus/immunology , Ileus/pathology , Macrophages/immunology , Monocytes/immunology , Postoperative Complications/immunology , Postoperative Complications/pathology , Receptors, CCR2/immunology , Animals , Cell Differentiation , Cell Movement , Disease Models, Animal , Gastrointestinal Motility , Gastrointestinal Transit , Homeostasis/immunology , Inflammation/immunology , Inflammation/pathology , Mice , Muscle, Smooth/pathology
13.
Mol Med ; 22: 464-476, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27341335

ABSTRACT

Vagotomy (VGX) increases the susceptibility to develop colitis suggesting a crucial role for the cholinergic anti-inflammatory pathway in the regulation of the immune responses. Since oral tolerance and the generation of regulatory T cells (Tregs) are crucial to preserve mucosal immune homeostasis, we studied the effect of vagotomy and the involvement of α7 nicotinic receptors (α7nAChR) at the steady state and during colitis. Therefore, the development of both oral tolerance and colitis (induced by dextran sulfate sodium (DSS) or via T cell transfer) was studied in vagotomized mice and in α7nAChR-/- mice. VGX, but not α7nAChR deficiency, prevented oral tolerance establishment. This effect was associated with reduced Treg conversion in the lamina propria and mesenteric lymphnodes. To the same extent, vagotomized mice, but not α7nAChR-/- mice, developed a more severe DSS colitis compared with control mice treated with DSS, associated with a decreased number of colonic Tregs. However, neither VGX nor absence of α7nAChR in recipient mice affected colitis development in the T cell transfer model. In line, deficiency of α7nAChR exclusively in T cells did not influence the development of colitis induced by T cell transfer. Our results indicate a key role for the vagal intestinal innervation in the development of oral tolerance and colitis, most likely by modulating induction of Tregs independently of α7nAChR.

14.
Nat Commun ; 15(1): 6079, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030280

ABSTRACT

Enteric glia have been recently recognized as key components of the colonic tumor microenvironment indicating their potential role in colorectal cancer pathogenesis. Although enteric glia modulate immune responses in other intestinal diseases, their interaction with the colorectal cancer immune cell compartment remains unclear. Through a combination of single-cell and bulk RNA-sequencing, both in murine models and patients, here we find that enteric glia acquire an immunomodulatory phenotype by bi-directional communication with tumor-infiltrating monocytes. The latter direct a reactive enteric glial cell phenotypic and functional switch via glial IL-1R signaling. In turn, tumor glia promote monocyte differentiation towards pro-tumorigenic SPP1+ tumor-associated macrophages by IL-6 release. Enteric glia cell abundancy correlates with worse disease outcomes in preclinical models and colorectal cancer patients. Thereby, our study reveals a neuroimmune interaction between enteric glia and tumor-associated macrophages in the colorectal tumor microenvironment, providing insights into colorectal cancer pathogenesis.


Subject(s)
Colorectal Neoplasms , Neuroglia , Signal Transduction , Tumor Microenvironment , Animals , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Humans , Tumor Microenvironment/immunology , Neuroglia/metabolism , Mice , Macrophages/metabolism , Macrophages/immunology , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Interleukin-6/metabolism , Monocytes/metabolism , Monocytes/immunology , Mice, Inbred C57BL , Cell Communication , Cell Differentiation , Cell Line, Tumor , Female
15.
Nat Commun ; 14(1): 509, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36720873

ABSTRACT

Spatially resolved transcriptomics has enabled precise genome-wide mRNA expression profiling within tissue sections. The performance of methods targeting the polyA tails of mRNA relies on the availability of specimens with high RNA quality. Moreover, the high cost of currently available spatial resolved transcriptomics assays requires a careful sample screening process to increase the chance of obtaining high-quality data. Indeed, the upfront analysis of RNA quality can show considerable variability due to sample handling, storage, and/or intrinsic factors. We present RNA-Rescue Spatial Transcriptomics (RRST), a workflow designed to improve mRNA recovery from fresh frozen specimens with moderate to low RNA quality. First, we provide a benchmark of RRST against the standard Visium spatial gene expression protocol on high RNA quality samples represented by mouse brain and prostate cancer samples. Then, we test the RRST protocol on tissue sections collected from five challenging tissue types, including human lung, colon, small intestine, pediatric brain tumor, and mouse bone/cartilage. In total, we analyze 52 tissue sections and demonstrate that RRST is a versatile, powerful, and reproducible protocol for fresh frozen specimens of different qualities and origins.


Subject(s)
RNA , Transcriptome , Child , Male , Humans , Animals , Mice , Transcriptome/genetics , RNA, Messenger , Benchmarking , Biological Assay
16.
Nat Rev Gastroenterol Hepatol ; 20(9): 597-614, 2023 09.
Article in English | MEDLINE | ID: mdl-37258747

ABSTRACT

The number of studies investigating the human gastrointestinal tract using various single-cell profiling methods has increased substantially in the past few years. Although this increase provides a unique opportunity for the generation of the first comprehensive Human Gut Cell Atlas (HGCA), there remains a range of major challenges ahead. Above all, the ultimate success will largely depend on a structured and coordinated approach that aligns global efforts undertaken by a large number of research groups. In this Roadmap, we discuss a comprehensive forward-thinking direction for the generation of the HGCA on behalf of the Gut Biological Network of the Human Cell Atlas. Based on the consensus opinion of experts from across the globe, we outline the main requirements for the first complete HGCA by summarizing existing data sets and highlighting anatomical regions and/or tissues with limited coverage. We provide recommendations for future studies and discuss key methodologies and the importance of integrating the healthy gut atlas with related diseases and gut organoids. Importantly, we critically overview the computational tools available and provide recommendations to overcome key challenges.


Subject(s)
Gastrointestinal Tract , Organoids , Humans , Forecasting
17.
Nat Biotechnol ; 40(4): 476-479, 2022 04.
Article in English | MEDLINE | ID: mdl-34845373

ABSTRACT

Current methods for spatial transcriptomics are limited by low spatial resolution. Here we introduce a method that integrates spatial gene expression data with histological image data from the same tissue section to infer higher-resolution expression maps. Using a deep generative model, our method characterizes the transcriptome of micrometer-scale anatomical features and can predict spatial gene expression from histology images alone.


Subject(s)
Transcriptome , Transcriptome/genetics
18.
Mucosal Immunol ; 15(6): 1296-1308, 2022 06.
Article in English | MEDLINE | ID: mdl-36071145

ABSTRACT

Monocyte-derived macrophages (Mφs) are crucial regulators during muscularis inflammation. However, it is unclear which micro-environmental factors are responsible for monocyte recruitment and anti-inflammatory Mφ differentiation in this paradigm. Here, we investigate Mφ heterogeneity at different stages of muscularis inflammation and determine how environmental cues can attract and activate tissue-protective Mφs. Results showed that muscularis inflammation induced marked alterations in mononuclear phagocyte populations associated with a rapid infiltration of Ly6c+ monocytes that locally acquired unique transcriptional states. Trajectory inference analysis revealed two main pro-resolving Mφ subpopulations during the resolution of muscularis inflammation, i.e. Cd206+ MhcIIhi and Timp2+ MhcIIlo Mφs. Interestingly, we found that damage to the micro-environment upon muscularis inflammation resulted in EGC activation, which in turn stimulated monocyte infiltration and the consequent differentiation in anti-inflammatory CD206+ Mφs via CCL2 and CSF1, respectively. In addition, CSF1-CSF1R signaling was shown to be essential for the differentiation of monocytes into CD206+ Mφs and EGC proliferation during muscularis inflammation. Our study provides a comprehensive insight into pro-resolving Mφ differentiation and their regulators during muscularis inflammation. We deepened our understanding in the interaction between EGCs and Mφs, thereby highlighting pro-resolving Mφ differentiation as a potential novel therapeutic strategy for the treatment of intestinal inflammation.


Subject(s)
Macrophages , Monocytes , Humans , Inflammation , Neuroglia , Anti-Inflammatory Agents
19.
Front Med (Lausanne) ; 8: 694268, 2021.
Article in English | MEDLINE | ID: mdl-34307422

ABSTRACT

Background: We previously showed increased susceptibility to dextran sulfate sodium (DSS)-induced colitis in vagotomized mice. Here, we evaluated whether vagus nerve stimulation (VNS) is able to reduce the severity of DSS colitis and aimed to unravel the mechanism involved. Methods: Colitis was induced in wild type mice by 2.5% DSS administration in drinking water for 5 days. VNS (5 Hz, 1 ms, 1 mA) was applied 1 day prior to and after 4 days of DSS administration to evaluate changes in epithelial integrity and inflammatory response, respectively. Epithelial integrity was assessed using TUNEL and Ki67 staining. Monocytes, immature and mature macrophages were sorted from colonic samples and gene expression levels of pro-inflammatory cytokines were studied. Results: VNS applied prior to DSS administration (i.e., prophylactic VNS) reduced disease activity index (VNS 0.8 ± 0.6 vs. sham 2.8 ± 0.7, p < 0.001, n = 5) and tended to improve histology score. Prophylactic VNS significantly increased epithelial cell proliferation and diminished apoptosis compared to sham stimulation. VNS applied at day 4 during DSS administration (i.e., therapeutic VNS) decreased the influx of monocytes, monocyte-derived macrophages and neutrophils, and significantly reduced pro-inflammatory cytokine expression (i.e., Tnfα and Cxcl1) in immature macrophages compared to sham stimulation. Conclusions: A single period of VNS applied prior to DSS exposure reduced DSS-induced colitis by an improvement in epithelial integrity. On the other hand, VNS applied during the inflammatory phase of DSS colitis reduced cytokine expression in immature macrophages. Our data further underscores the potential of VNS as novel therapeutic approach for inflammatory bowel diseases.

20.
Curr Opin Neurobiol ; 62: 68-75, 2020 06.
Article in English | MEDLINE | ID: mdl-31862627

ABSTRACT

Intestinal homeostasis relies on the reciprocal crosstalk between enteric neurons and immune cells, which together form neuro-immune units that occupy distinct anatomical niches within the gut. Here we will review the recent advances in our understanding of neuro-immune crosstalk within the gut, with focus on macrophages, mast cells and innate lymphoid cells. In particular, we will discuss the role of neuron-immune cell crosstalk in homeostasis, and how aberrant communication may underlie disease in the gastro-intestinal tract.


Subject(s)
Lymphocytes , Macrophages , Mast Cells , Immunity, Innate , Intestinal Mucosa
SELECTION OF CITATIONS
SEARCH DETAIL