Next Article in Journal
Identification of Aging and Young Subtypes for Predicting Colorectal Cancer Prognosis and Immunotherapy Responses
Next Article in Special Issue
CD4 T-Cell Subsets and the Pathophysiology of Inflammatory Bowel Disease
Previous Article in Journal
Human Regulatory T Cells: Understanding the Role of Tregs in Select Autoimmune Skin Diseases and Post-Transplant Nonmelanoma Skin Cancers
Previous Article in Special Issue
Targeting Lineage-Specific Transcription Factors and Cytokines of the Th17/Treg Axis by Novel 1,3,4-Oxadiazole Derivatives of Pyrrolo[3,4-d]pyridazinone Attenuates TNBS-Induced Experimental Colitis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathophysiology of Inflammatory Bowel Disease: Innate Immune System

by
Angela Saez
1,2,†,
Beatriz Herrero-Fernandez
1,3,†,
Raquel Gomez-Bris
1,3,
Hector Sánchez-Martinez
1 and
Jose M. Gonzalez-Granado
1,4,5,6,*
1
LamImSys Lab, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
2
Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), 28223 Pozuelo de Alarcón, Spain
3
Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
4
Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
5
CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
6
Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(2), 1526; https://doi.org/10.3390/ijms24021526
Submission received: 13 December 2022 / Revised: 30 December 2022 / Accepted: 10 January 2023 / Published: 12 January 2023

Abstract

:
Inflammatory bowel disease (IBD), comprising Crohn’s disease (CD) and ulcerative colitis (UC), is a heterogeneous state of chronic intestinal inflammation with no exact known cause. Intestinal innate immunity is enacted by neutrophils, monocytes, macrophages, and dendritic cells (DCs), and innate lymphoid cells and NK cells, characterized by their capacity to produce a rapid and nonspecific reaction as a first-line response. Innate immune cells (IIC) defend against pathogens and excessive entry of intestinal microorganisms, while preserving immune tolerance to resident intestinal microbiota. Changes to this equilibrium are linked to intestinal inflammation in the gut and IBD. IICs mediate host defense responses, inflammation, and tissue healing by producing cytokines and chemokines, activating the complement cascade and phagocytosis, or presenting antigens to activate the adaptive immune response. IICs exert important functions that promote or ameliorate the cellular and molecular mechanisms that underlie and sustain IBD. A comprehensive understanding of the mechanisms underlying these clinical manifestations will be important for developing therapies targeting the innate immune system in IBD patients. This review examines the complex roles of and interactions among IICs, and their interactions with other immune and non-immune cells in homeostasis and pathological conditions.

1. Introduction

Inflammatory bowel disease (IBD) encompasses a group of heterogeneous diseases that entail chronic, relapsing gastrointestinal tract inflammation of inexactly known etiology and pathogenesis. IBD etiology may involve the host immune system, genetic variability, and environmental factors [1]. IBD is clinically classified as Crohn’s disease (CD) or ulcerative colitis (UC) based on symptoms, disease location, and histopathological characteristics. UC causes long-lasting inflammation and superficial ulcerative disease in the colon, whereas CD is a transmural disease often associated with granuloma formation and can appear in any part of the gastrointestinal tract [2,3,4,5]. IBD can be associated with life-threatening conditions, including primary sclerosing cholangitis, blood clots, and colon cancer [6]. IBD is usually diagnosed between the ages of 20 and 40 years, but can start at any age. IBD shows alternating periods of clinical relapse and remission.
The intestinal mucosa is composed of epithelial cells, goblet and Paneth cells, stroma and immune cells. The intestinal epithelium includes a monolayer of epithelial cells closely bound by tight junctions and interposed with immune cells. The intestine is structured as a series of protrusions known as villi and invaginations called crypts of Lieberkühn [7]. The epithelium participates in nutrient absorption and, at the same time, interposes a physical barrier to the contents of the intestinal lumen. The epithelium also interacts with the intestinal microbiota and the immune system, sending receiving signals to and from both.
The epithelium includes goblet and Paneth cells, which, respectively, produce mucus and antimicrobial peptides, thus limiting the spread of luminal microorganisms [7]. A marked reduction in goblet cell numbers has been linked to a loss of mucus layer thickness in Crohn’s disease [8], and abnormal mucus composition has been reported in UC [9]. Beneath the epithelium, the lamina propria contains stromal cells, including fibroblasts, myofibroblasts, and perivascular pericytes. These cells serve functions in fibrosis and wound healing [7], and may be related to the aggravation of UC through their capacity to produce chemokines, including chemokine (C-C motif) ligand (CCL)19, CCL21, and the immune-system regulator interleukin (IL)-33 [10].
Plasma cells release immunoglobulin (Ig)A, inhibiting the infiltration of pathogenic microorganisms and helping to sustain a homeostatic equilibrium between the host and commensal microbiota. Both the epithelium and other non-immune intestinal components are important mediators of intestinal homeostasis and IBD pathophysiology, reviewed in [11,12]. However, some of the functions of these non-immune cells are mediated through interaction with components of the immune system, as will be described in this review.
The immune system confers host defense against pathogens and provides anti-tumor protection. At the same time, regulatory mechanisms counterbalance these responses to prevent reactions against self and innocuous external antigens, thus promoting a state of tolerance.
The immune system can be classified into innate and adaptive immunity. Innate immunity, composed of myeloid cells among other elements, initiates rapid and nonspecific responses to conserved structural motifs on microorganisms. Innate immune cells (IIC) express pattern recognition receptors (PRRs), such as toll-like receptors (TLRs) and Nod-like receptors (NLR), allowing them to distinguish pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). IIC promote host defense and inflammation by generating cytokines and chemokines, triggering the complement cascade and phagocytosis, or stimulating adaptive immunity by presenting antigens. Notable IIC include neutrophils, monocytes, macrophages, and dendritic cells (DCs) [13,14].

2. IBD Pathophysiology

The etiology of IBD remains elusive, but IBD appears to be sustained in genetically susceptible individuals by an impaired immune response against intestinal microorganisms. This abnormal immune response is associated with dysregulation of both innate and adaptive immune responses.
IBD features breach the epithelial barrier of specific zones in the intestine, and non-resolving mucosal damage is thought to be an important characteristic of the disease [15]. While generally unknown, the cause of this damage could be related to an infectious agent [16], a chemical compound [1], or a metabolic alteration probably related to diet-mediated dysbiosis [17]. The disease is then thought to be perpetuated by deficient resolution of the inflammatory response to this initial injury [18]. Unsuccessful resolution of inflammation is possibly supported by disruption of tolerance towards commensal microorganisms or autologous signals of tissue damage [15,19].
There is also some uncertainty as to whether the epithelial barrier alterations precede or follow the development of inflammation in the lamina propria [20].

2.1. Innate Immune Cells in the Pathogenesis of IBD

In IBD, the innate immune system is the first responder to PAMPs and to molecules released from damaged or dying cells, known as DAMPs. DAMPs and PAMPs activate the innate immune system by interacting with PRRs. These patterns can be sensed by several components of the innate immune system, including granulocytes, neutrophils, monocytes, myeloid-derived suppressor cells, macrophages, and dendritic cells. In addition, these patterns can also be recognized by non-immune cells, such as intestinal epithelial cells (IECs) and myofibroblasts.
IICs respond to these signals, temporarily enhance the epithelial barrier, and clean up the effects of inflammation [21]. The implication of IIC in IBD is the focus of this review.

2.1.1. Neutrophils in Gut Homeostasis

Neutrophils are the most numerous immune cells in the human circulation and are quickly recruited to sites of infection or inflammation [22], forming the first line of immune defense.
When the intestinal barrier is damaged, neutrophils are recruited from the circulation to the inflamed tissue through a plethora chemotactic gradients formed by cytokines such as IL-1β, IL-6, and tumor necrosis factor (TNF)-α; chemokines such as CCL8, chemokine (C-X-C motif) ligand (CXCL)10, and macrophage inflammatory protein 2 (MIP)-2 (also known as CXCL2); and growth factors such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte colony-stimulating factor (G-CSF) [23,24,25]. Neutrophil recruitment is also mediated by bacteria-derived molecules such as formyl-methionyl-leucyl-phenylalanine and short-chain fatty acids (SCFAs) [26,27,28] (Figure 1).
Neutrophils participate in the elimination of microorganisms through phagocytosis, degranulation, the generation of reactive oxygen species (ROS), and the release of neutrophil extracellular traps (NETs). NETs are mesh-like structures made of DNA and its histone scaffold together with granule components such as myeloperoxidase (MPO), cathepsin G, neutrophil elastase, and protease 3. NETs protrude from the membrane of the activated neutrophil to restrain large microorganisms, activate complement factors, and therefore facilitate contained lysis through their bactericidal and permeability-increasing actions [29,30,31,32]. Once their functions are completed, neutrophils undergo apoptosis and efferocytosis, facilitating resolution of the inflammatory response, repair, and a return to normal tissue homeostasis [33,34,35].

2.1.2. Neutrophils in the Gut during IBD

Some studies have described the participation of NETs in IBD as a double-edged sword. On the one hand, NETs can impede the spread of microorganisms by trapping them in an environment of microbicidal components, while also stimulating the healing of the intestinal mucosa upon injury and helping to sustain the stability of the intestinal epithelium [36,37]. On the other hand, increased neutrophil activity and exacerbated NETs production can impair intestinal mucosal barrier function, damage the intestinal epithelium, and accentuate disease symptoms [38]. Neutrophils directly promote tissue damage by releasing proteases such as matrix metalloproteinases (MMPs) and neutrophil elastase, and by altering membrane properties by releasing ROS [39] (Figure 1).
Proteomics studies and microscopy validations have identified eleven neutrophil- and NETs-associated proteins with increased abundance in biopsies from UC patients [40], and similar results have been reported by others [39].
Dextran sodium sulfate (DSS)-induced colitis in mice promotes the accumulation of NETs in the colon, accompanied by the induction of epithelial cell death by apoptosis, breakage of tight junctions, increased permeability, and augmented bacterial translocation and inflammation [41]. Moreover, NETs accumulation boosts TNF-α and IL-1β production in plasma by signaling via the ERK1/2 pathway. The reduction of NETs protects against colitis and inhibits the augmentation of pro-inflammatory factors implicated in IBD [41].
An increase in neutrophil activity has been observed in IBD patients [42]. This increase is associated with the release of TNF-α and the presence of lipopolysaccharides, two factors that contribute to neutrophil activation. Other factors that can contribute to NETs production are IL-8 produced by endothelial cells, and an increased abundance of protein arginine deiminase 4 (PAD4) [30,31,43]. PAD4 is more abundant in intestinal tissue from UC patients than healthy individuals [31], and in damaged tissue rather than healthy tissue from the same individual [30,31,32]. PAD4 mediates histone citrullination, an important event in NETosis that precedes chromatin decondensation and DNA release.
An analysis of NETs-associated proteins in colon samples from patients with UC, CD, and colon cancer showed a greater abundance of PAD4, MPO, neutrophil elastase, and citrullination histone H3 (CitH3) in UC than in CD. Moreover, neutrophils in UC tissues produced more NETs upon treatment with TNF-α [44]. In a similar way, increased expression of Ly6G, CitH3, and PAD4 has been found in mouse colon in a model of colitis induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS) [45]. This effect was associated with damage to the intestinal epithelial barrier [45]. Reduced NETs formation ameliorated colitis symptoms and tissue damage [46,47,48,49].
Neutrophils promote IBD gut inflammation by producing high levels of ROS that impair the epithelial barrier and promote redox-sensitive inflammatory pathways [50]. The epithelial barrier is also damaged by neutrophil-produced proteases, pro-inflammatory cytokines such as IL-8, TNF-α, and leukotriene B4, which additionally recruit monocytes and more neutrophils to the inflamed tissue [51,52].
Another cytokine recently proposed to promote neutrophil recruitment to colonic tissue is IL-22, whose levels correlate with neutrophil infiltration [53]. Neutrophils can be activated by the cytokines IL-1β and IL-18 [54], produced during inflammasome assembly [55,56], and upon the release by necrotic cells of the nuclear ‘alarmin’ IL-1α [57]. Other mediators of neutrophil action altered in IBD include GM-CSF and G-CSF [58] and IL-17A and IL-17F [59], which act through the IL-23–IL-17A–G-CSF axis [60,61], providing a possible explanation for the continuous regeneration of neutrophils in IBD [15]. Neutrophils also express the IBD-protective gene caspase recruitment domain 9 (Card9), which provides them with the capacity to protect against DSS-induced colitis; a lack of CARD9 enhances mitochondrial dysfunction and ROS generation, leading to neutrophil apoptosis and increased inflammation [62].
The intestinal epithelium constitutes a physical barrier that isolates subepithelial tissues from luminal contents, providing a fundamental support for intestinal homeostasis. Neutrophils are associated with bystander tissue damage, but also play a role in epithelial restitution [63].
As well as killing microorganisms, neutrophils cooperate in wound healing and the resolution of inflammation by releasing vascular endothelial growth factors and beneficial lipid mediators such as protectin D1 and resolvin E1. These factors impede further neutrophil recruitment and augment the phagocytosis of apoptotic neutrophils by macrophages. Neutrophils also remove cellular debris from sites of inflammation by phagocytosis [61,64,65,66]. The release of proteases within NETs can regulate cytokine function by proteolysis [67], and this phenomenon might also regulate cytokines in IBD.
Contrasting these protective effects, the accumulation of hyperactivated neutrophils promotes an alteration of crypt structure and the formation crypt abscesses. This process features a disproportionate enzymatic reaction, generation of the pro-inflammatory cytokines TNF-α and IL-1β, and the secretion of the non-cytokine inflammatory molecules α defensins and calprotectin, attracting monocytes, T cells, and more neutrophils to the inflammation site and promoting the pathogenesis of IBD [68,69,70,71,72]. Neutrophils can also promote goblet-cell depletion, a main feature of IBD [73,74,75].
Neutrophils are also, themselves, regulated by the effects of intestinal epithelial cells during intestinal inflammation, a topic recently reviewed in [63].
In summary, neutrophils play intricate roles in intestinal inflammation, contributing to the elimination of invading pathogens and epithelial restitution, while at the same time participating in the disruption of crypt architecture and generating bystander tissue damage, roles that impede and promote the development of the IBD, respectively.
Under the influence of neutrophils, other phagocytic cells such as monocytes and macrophages remove cell debris and help to distinguish damaged areas from tissue areas less affected by acute inflammation [76].

2.1.3. Macrophages in the Gut in Steady State Conditions

Macrophages are highly plastic cells, and their functions depend on their developmental ontogeny and surrounding environment [77,78,79] (Figure 2). During embryonic development, self-maintaining tissue-resident macrophages derive from the yolk sac and fetal liver progenitors [80,81,82,83]. After birth, blood circulating bone marrow-derived monocytes are recruited to tissues, where they differentiate to macrophages, replenishing tissue-resident populations and adopting a phenotype conditioned by the local tissue environment [82,84,85,86].
Both tissue-resident and monocyte-derived macrophages perform the typical macrophage functions of phagocytosis, cytokine production, and host interaction [87]. In the intestine, monocyte-derived macrophages are more abundant than macrophages of embryonic origin [77]; however, distinct subsets of embryonic-derived macrophages remain in the intestine even after weaning [88].
Upon weaning, bone marrow-derived monocytes egress from the circulation and extravasate into the tissue, contributing to macrophage replenishment in the intestine [89]. After extravasation, monocytes undergo differentiation and maturation processes, acquiring a more macrophage-like phenotype. These changes include downregulation of Ly6C generation, production of class II major histocompatibility complex molecules (MHCII), and increased expression of C-X3-C motif chemokine receptor1 (CX3CR1); monocyte-to-macrophage transition thus involves a switch from a Ly6Chigh, CX3CR1int state to a mature Ly6C, CX3CR1high, CD64+, MHCII+ state [77,90,91].
In steady-state conditions, intestinal macrophages are distributed throughout the gut structure, including the lamina propria, submucosa and the muscularis externa [92].
Distal to the gut lumen, in the muscularis externa, are located long-lived, bipolar, and stellate self-renewing embryonic-derived macrophages [93]. Muscularis macrophages interact with enteric and myenteric neurons, influencing enteric neurons [94], and controlling intestinal functions including secretion and motility [95,96,97].
The macrophages of the lamina propria are rounded monocyte-derived cells that are constantly replenished [89,94]. These macrophages are short lived and are distributed close to the lumen, where they constantly encounter the gut microbiota [93,98] and contribute to oral tolerance [99,100]. In this layer, macrophages provide signals to the intestinal stem cells, which give rise to goblet cells, Paneth cells, and intestinal epithelial cells [101,102]. These macrophages also modulate regulatory T cell (Treg) activity and function via the secretion of IL-10 [103] and T helper (Th)17 cells by providing IL-1β [104]. Lamina propria macrophages thus support intestinal homeostasis through a mix of phagocytic and antibacterial functions, immune modulation, and tissue repair [105,106].
Intestinal-tissue macrophage phenotypes and functions depend on the microbiota and their metabolites and on microenvironmental cues [77,107,108,109]. Intestinal macrophages can thus be classified into three distinct populations according to their homeostatic function: host defense, wound healing, and immune regulation [110].
Host defense macrophages have microbicidal activity and, upon stimulation with interferon (IFN)γ or TNF-α, generate cytokines that are commonly secreted by T cells, natural killer cells, or antigen presenting cells (APC). Wound healing macrophages develop upon contact with IL-4 released by T cells or granulocytes, and play a role in tissue repair. Regulatory macrophages, which have an anti-inflammatory function, are generated in response to several stimuli, including IL-10, glucocorticoids, and apoptotic cells [110].
Intestinal macrophages need to combat invading pathogenic bacteria while tolerating beneficial probiotic bacteria [111]. To discern between commensal and harmful bacteria, macrophages recognize harmful bacteria through PRRs such as TLRs and NOD-like receptors (NLRs) [110,112,113]. Intestinal macrophages are excellent phagocytes of harmful bacteria, but upon engulfing or recognizing harmful bacteria they produce low amounts of pro-inflammatory cytokines [114,115,116,117]. In contrast, they naturally produce elevated amounts of IL-10, which is associated with their reduced response to TLR-triggering [103,114,118,119,120,121,122], suggesting a somewhat anti-inflammatory phenotype for resident intestinal macrophages, which avoid bacteria-activated inflammation in the gut under steady state conditions [117,123].
In the gut, IL-34 and CSF-1 (also called M-CSF)—both ligands for the CSF-1 receptor (CSF-1R)—promote monocyte and macrophage differentiation [124,125], and mice lacking CSF-1 or CSF-1R are deficient for tissue macrophages [126,127,128,129]. Supporting this, administration of anti-CSFR antibodies reduces macrophage numbers [130], while recombinant CSF-1 increases intestinal macrophage infiltration [131].
Intestinal macrophages are involved in phagocytosis and the clearance of apoptotic cells [132,133], including apoptotic IECs, helping to maintain epithelial barrier integrity under steady state conditions [134].
CX3CR1high intestinal macrophages sense and take up bacterial antigens from the intestinal lumen through their transepithelial dendrites [135,136,137,138,139,140]. In homeostasis, intestinal microbiota inhibit the migration of antigen-loaded CX3CR1high intestinal macrophages to mesenteric lymph nodes, thereby also inhibiting antigen presentation to T cells and effectively sustaining tolerance towards commensal bacteria. When the intestinal microbiota is disturbed or under chronic colitis conditions, CX3CR1high macrophages can change their habits and migrate to lymph nodes [121].
Intestinal macrophages also regulate other immune cells. CX3CR1high macrophages capture soluble food antigens and transfer them to CD103+ dendritic cells, promoting antigen presentation and food tolerance [141,142]. Lamina propria macrophages produce IL-10, which promotes the differentiation of Forkhead Box P3 (Foxp3)+ Tregs [103,118,143], and also produce IL-1β, which acts on Th17 cells [104]. Intestinal macrophages also produce IL-1α and IL-1β in response to commensal microbiota, affecting Group 3 innate lymphoid cells (ILC3), and GM-CSF, which acts on macrophages and dendritic cells to maintain Treg homeostasis [107]. CX3CR1+ mononuclear phagocytes prime T cells and promote Th17 cell differentiation [144]. Taken together, these observations show that intestinal macrophages and their secreted cytokines regulate T cell responses in the gut.

2.1.4. Macrophages in the Pathogenesis of IBD

In IBD, the local release of PAMPs and DAMPs at the site of injury triggers intestinal inflammation. During this process, large numbers of Ly6Chigh inflammatory monocytes are recruited to the intestinal tissue in a process dependent on C-C motif chemokine ligand 2 (CCR2) [89,90,145,146], which is also known as monocyte chemoattractant protein (MCP)-1. Lack of CCR2 in mice abrogates the recruitment of TLR2+ CCR2+ Gr-1+, TNF-α-producing macrophages to the inflamed intestine [147], and reduces symptoms of DSS-induced colitis [147,148] (Figure 2).
Monocyte migration to the lamina propria is also controlled by IL-8 and transforming growth factor (TGF)-β, constitutively generated by mucosal epithelial cells [149].
Mouse models of IBD reveal significantly elevated numbers of macrophages in the colon, characterized by an increase in the proportion of Ly6C+ macrophages relative to mature Ly6C macrophages. This Ly6C+ macrophage recruitment is dependent on the expression of CCR2 [90,147]. Interestingly, a pronounced elevation in colonic macrophage numbers, and an increased proportion of Ly6C+MHCII+ monocyte-derived macrophages are key features of spontaneous colitis in IL-10R-deficient mice on the C57BL/6 background [150].
IBD patients also have an increased number of pro-inflammatory macrophages [90,151,152], and pediatric IBD patients show accumulation of activated mucosal macrophages [153]. Usually, these macrophages have augmented expression of pro-inflammatory molecules such as TNF-α, IL-1β, IL-6, and inducible nitric oxide synthase (iNOS) [151,154]. Recruited Ly6Chigh monocytes in IBD upregulate TLR2 and NOD2, which increases their sensitivity to bacteria and triggers their differentiation to pro-inflammatory effector cells [122]. These inflammatory macrophages produce TNF-α, IL-6, and iNOS [110,155]. Resident CX3CR1high macrophages maintain their anti-inflammatory phenotype even when sharing the intestine with Ly6Chigh inflammatory macrophages [90,116].
In steady state conditions, CX3CR1high resident macrophages, which are highly phagocytic and MHCIIhigh but resilient to TLR-stimulation and constitutively IL-10 producers, are accompanied by a small population of CX3CR1int cells, mainly resulting in a CX3CR1high resident macrophage population. CX3CR1int cells give rise to CX3CR1high macrophage. In IBD, this CX3CR1int to CX3CR1high macrophage conversion is diminished, leading to the accumulation of TLR-reactive inflammatory CX3CR1int macrophages [90].
CX3CR1 and its ligand CX3CL1 are upregulated in the colon of IBD mice and seem to play an important role in the disease [156], with CX3CR1 and CX3CL1 polymorphisms in patients linked to the clinical manifestations of IBD [157,158]. However, it is unclear if they play a protective or harmful role, since their deficiency protects from [138] or aggravates [156] experimentally induced colitis depending on the study.
Macrophage-expressed IL-10 and its receptor IL-10R support intestinal homeostasis and are implicated in the development of IBD [159,160,161,162,163,164,165]. IL-10–IL-10R-signaling mediates the differentiation and function of intestinal macrophages in mice and IBD patients [166]. The absence of IL-10 in mice provokes a shift from the resident CX3CR1high macrophage phenotype in the colon to a pro-inflammatory phenotype [121]. Interestingly, specific depletion of IL-10 in CX3CR1high intestinal macrophages has no effect on intestinal homeostasis or Treg regulation [121], but the intestinal macrophage-specific lack of IL-10R alters intestinal homeostasis and provokes severe gut inflammation [121,167]. The absence of Il-10ra in intestinal macrophages promotes the production of IL-23, which in turn mediates IL-22 secretion by Th17 and ILC3 cells. IL-22 activates IECs to express an antimicrobial peptide that induces neutrophil recruitment, promoting IBD [167]. Moreover, colitis symptoms are ameliorated by manipulation of the microbiota to increase IL-10 producing macrophage numbers [109].
Lamina propria macrophages produce the chemokine CCL8, recruiting circulating Ly6Chigh monocytes in IBD [122,168].
Intestinal macrophages are also important for epithelial tissue repair. These macrophages produce molecules that control epithelial regeneration [169]. As mentioned, intestinal macrophages in the pericryptal stem cell niche stimulate neighboring colonic epithelial progenitors and promote epithelial recuperation after injury [170]. Epithelial recovery is also stimulated by intestinal macrophage-derived IL-10, through the stimulation of the CREB/WISP-1 pathway in epithelial cells [171,172] or by macrophage production of hepatic growth factor (HGF). Curiously, CD-patient-derived macrophages show diminished HGF secretion, possibly affecting epithelial restoration in these patients [173]. Classically, two main polarized macrophage phenotypes have been proposed, pro-inflammatory M1 and anti-inflammatory M2 [174]. Tissue repairing M2-like macrophages promote stimulation of wingless-related integration site (WNT) signaling in response to differentiation driven by signal transducer and activator of transcription (STAT)-6 [175] through a mechanism dependent on the alarmin IL-33 [176]. STAT6-dependent M2-like macrophage differentiation promotes the stimulation of WNT signaling to promote tissue repair [175]. The alarmin IL-33 also favors protective M2-like macrophages polarization and subsequent mucosal repair [176,177]. Another study showed that altered α4β7-mediated intestinal-homing of non-classical monocytes might reduce the number of wound-healing macrophages, leading to impaired intestinal wound healing [178].
Upon tissue injury, macrophages are activated to combat microbiota invasion through phagocytosis and to facilitate repair of the damaged tissue. However, when macrophages are improperly activated, they directly cause the onset and development of fibrosis [179,180]. Fibrosis is a disproportionate accumulation of extracellular matrix (ECM) components such as collagen [181]. Excessive fibrosis produces a non-optimal tissue architecture, and intestinal fibrosis is a common problematic characteristic of IBD [182,183]. Advanced intestinal fibrosis frequently results in intestinal strictures [106]. Macrophages promote myofibroblast-mediated fibrosis by producing TGF-β1, connective tissue growth factor (CTGF), and fibroblast activation protein (FAP). They also undergo macrophage-to-myofibroblast transition (MMT), favoring myofibroblast accumulation and excess ECM production [106]. In contrast, intestinal macrophages can restrict intestinal fibrosis by promoting myofibroblast senescence, degrading the ECM, and clearing profibrotic components [184]. Macrophages are not the only source of gut fibrosis in IBD; IL-34, which is overproduced in IBD and mediates macrophage maturation [125], also activates collagen synthesis by gut fibroblasts [185].
In summary, macrophages play an important role in homeostasis and in the development of IBD [121,150,166,167], both in mouse models and in patients, by phagocytosing cellular debris, producing multiple cytokines, and regulating tissue repair [105].

2.1.5. Innate Lymphoid Cells in IBD

Another important component of the innate immune system in the intestine is the population of innate lymphoid cells (ILCs). ILCs are important mediators of antimicrobial defense and contribute to organ development, tissue protection and regeneration, and mucosal homeostasis [186,187]. These cells belong to the innate immune system, but are derived from the same common lymphoid progenitor population as lymphocytes [188,189]. ILCs act early in the immune response by replying quickly to cytokines and other signals produced by other cells [188,190,191,192,193,194]. Recent discoveries have highlighted the essential role of ILCs in intestinal mucosal homeostasis and IBD [195,196,197,198,199,200]. However, they will be not discussed in this review since we and others have recently reviewed their role in IBD [194,201,202,203,204].

2.1.6. Dendritic Cells in Homeostasis

DCs link the innate and adaptive immune systems by presenting antigens to and activating T cells. Several DC subtypes are derived from a specific common dendritic progenitor (CDP). CDPs generate plasmacytoid DCs (pDCs) in bone marrow, as well as pre-DCs that circulate in the blood and give rise to conventional or classical DCs (cDCs) in lymphoid and nonlymphoid organs [205,206,207,208,209]. The production of pDCs versus cDCs is determined by the growth factor fms-like tyrosine kinase 3 ligand (Flt3L) [210] (Figure 2).
Monocytes originate in the bone marrow and circulate in blood. DCs can also originate from circulating monocytes after monocyte migration to inflamed tissues, where they differentiate to macrophages or monocyte-derived DCs through the action of M-CSF or GM-CSF, respectively. Monocyte-derived DCs belong to the mononuclear phagocyte system (MPS), and are better APCs than monocytes [211,212]. Through their function as professional APCs and their capacity to release cytokines, DCs play important roles in initiating immune responses to invading pathogens; cDCs and monocyte-derived DCs are powerful APCs, whereas pDCs specialize in secreting type I IFN [213]. Moreover, cDC subpopulations included chemokine (C motif) receptor 1 (XCR1)+ cDC1s and signal-regulatory protein alpha (SIPRα)+ cDC2s [205,206,207,208,209]. cDC2s can be subclassified as CD103+ or CD103. These DC subsets possess distinctive functional, phenotypical, and transcriptional features. While cDC1 are excellent APCs to cytotoxic T cells, cDC2s are more similar to pDCs in their capacity to polarize CD4+ T cell responses and to promote anti-viral responses via type I IFN [12].
Under physiological conditions, immature DCs patrol peripheral tissues, where they encounter and take up antigens [214]. Upon activation, maturing DCs increase their expression of MHCII and the costimulatory molecules CD80, CD86, and CD83 [209,213] and migrate along a chemokine gradient to draining lymph nodes, where they enter paracortical T cell zones to activate and prime antigen-specific naïve T cells and secrete cytokines [215,216,217,218]. In this way, DCs link innate and adaptive immunity by presenting antigens to and activating T cells.

2.1.7. Dendritic Cells in IBD

DCs accumulate in specific gut locations such as Peyer’s patches, isolated lymphoid follicles, and gut-associated lymphoid tissues. Like macrophages, DCs are constantly replenished from bone marrow-derived progenitors [88] (Figure 2).
Like some macrophages, DCs take up soluble food antigens directly from the intestinal lumen [136], but also take up food antigens from epithelial M-cells in the follicle-associated epithelium of Peyer’s patches [219]. As mentioned, DCs also receive antigens from CX3CR1high lamina propria macrophages through gap junctions [141,142]. In steady state, DCs play a tolerogenic role upon recognizing commensal bacterial components. XCR1+ cDC1s are important for intestinal homeostasis and in particular the expression of XCR1; mice lacking XCR1 in cDC1 lack intraepithelial and lamina propria T cell populations, and are more vulnerable to chemically-induced colitis [220].
CD103+ cDC2s seem to be important for initiating oral tolerance, in part through their capacity to generate retinoic acid (RA) required for the development of Foxp3+ Treg cells [221,222]. Furthermore, mammalian target of rapamycin (mTOR) protein kinase intervenes in the regulation of intestinal homeostasis by enhancing IL-10 production in cDC2s. Indeed, loss of mTOR signaling in DCs blocks IL-10 generation by cDC2s and increases sensitivity to DSS-induced colitis [223].
Colonic DCs display an abnormal immature phenotype in IBD that includes the expression of homing markers [224]. Intestinal DCs from UC patients have diminished expression of cutaneous lymphocyte antigen (CLA) and CCR4, while showing enhanced expression of CCR9 and β7 integrin [225,226].
DCs from CD-patient mucosa express more CD40 and release more IL-6 and IL-12 than DCs from healthy individuals [224]. In IBD, mucosal DCs show increased expression of TLR2 and TLR4 [225]. CD103+ CD11b+ cDCs are significantly reduced in abundance in the inflamed and uninflamed intestinal tissue of CD patients [227]. pDCs are also found in the inflamed gut, although their specific role is still undetermined [228].
The activation of intestinal CD103+ DCs in IBD patients results in the upregulation of PRRs. Thus, local variations in the gut microbiota may change the balance and regulation signals received by mucosal DCs. Upon activation, DCs are able to release inflammatory cytokines [229]. In summary, DCs play an essential role in IBD pathogenesis.
IBD pathogenesis can be augmented by inappropriate macrophage and DC responses to the microbiota [230]. These responses involve inadequate protection and strengthen pathogenicity.
Intestinal DCs promote tolerance to luminal antigens under physiological conditions, but can develop into an inflammatory response after inflammation or direct stimulation by TLR ligands. In these circumstances, intestinal DCs can release inflammatory cytokines such as IL-12, IL-6, and IL-18 and mediate Th1 responses when triggered. Supporting this, the circulation of IBD patients with active disease contains pDCs that migrate to secondary lymphoid organs, where they produce Th1 cytokines (IL-6, IL-8, and TNF-α), thereby perpetuating disease [230]. In addition, inflamed, and uninflamed intestine of CD patients has a reduced abundance of CD11c+ DCs, conferring an increased capacity to produce Th1/Th2/Th17 responses [231].
The role of cDC2s in IBD is less clear, with some studies pointing to an implication in T cell-mediated colitis [232,233], while others show no effect of lamina propria CD103+ cDC2s [233,234]. However, conditional absence of interferon regulatory factor (IRF)4 in mice results in abnormal development of colon lamina propria cDC2s and late initiation of T cell-dependent colitis [232], indicating a role of IRF4-expressing cDC2s in the initial priming of colitogenic T cells. Remarkably, cDC1s may play a protective role in the development of IBD, since lack of these cells increases predisposition to DSS-induced colitis [220,233].

3. Discussion

Comparison of first-degree relatives of IBD patients with the general population reveals a heritable risk of CD and UC [235,236]. Technological advances in genetic testing and DNA sequencing have allowed the development of genome-wide association studies (GWAS), which have identified more than 240 risk variants associated with IBD. These variants are found in genes related to bacteria recognition (e.g., NOD2), autophagy (e.g., ATG16L1 and IRGM), regulation of epithelial barrier (e.g., ECM1), and innate and adaptive immunity (e.g., IL-23R, IL-10, ITGAL, and ICAM1 variants) [237,238]. From these data, it has been possible to uncover fundamental molecular features underlying the disease and to identify genes and signaling pathways that represent potential therapeutic targets or biomarkers. However, only a small percentage of the disease variance in CD and UC can be linked to recognized IBD risk loci [239]. Most GWAS analyses in the field have examined whole intestinal tissue. One consequence of this is that that the data obtained reflect the most highly expressed mRNA transcripts in the more abundant cell populations, thus potentially missing less abundantly expressed genes. Moreover, the obtained data cannot be unambiguously associated with a specific cell population.
To resolve this limitation, new techniques have allowed the study of single-cell-specific transcriptional profiles. For example, single-cell RNA sequencing (scRNA-seq) and high-dimensional protein analyses, such as mass cytometry and multichannel spectral cytometry, have defined IBD-linked profiles and detected cell subpopulations that are elevated or diminished in IBD, particularly populations of fibroblasts [10], epithelial cells [9], and immune cells [240,241,242,243,244,245,246,247,248].
A complementary approach to GWAS is through transcriptome-wide association studies (TWAS), which associate gene expression with genetic susceptibility to disease, providing functional insight into risk loci [249]. TWAS findings have provided understanding of tissue-specific molecular events underlying genetic susceptibility to IBD. Associated genes are potential targets for new treatments and could be prioritized in functional studies.
Neutrophils play a dual role in intestinal homeostasis and inflammation, playing an essential role in gut defense but also, upon excessive recruitment, being an important mediator of tissue damage in the inflamed mucosa. Several studies demonstrate the effect of neutrophils on other components of the intestinal mucosa in IBD, such as other immune cells and epithelial cells and other non-immune cells. However, less is known about the reciprocal effect on neutrophils of these other cells, particularly intra-epithelial cell components as stroma and epithelial cells, although some studies point to the importance of neutrophil–epithelial cell communication for essential neutrophil functions such as recruitment, transepithelial migration, cell death, and clearance (Reviewed in [63]).
Pharmacological inhibition of NET formation ameliorates IBD [39]. Potentiation of the protective functions of neutrophils, in combination with a reduction in the adverse effects of NET formation, offers an interesting approach to the treatment of intestinal diseases; however, this regulation must be achieved without compromising neutrophil immune-defense functions. Targeting NETs is a potentially interesting approach to achieving complete mucosal healing in IBD. Another area of interest for therapeutic intervention is the development of markers of NET formation in vivo. Supporting this, calprotectin (s100A8/9), the most commonly used fecal biomarker in IBD, constitutes up to 60% of neutrophil cytosolic protein content [250,251]. Calprotectin is also found in NETs [36]. Other neutrophil-associated IBD biomarkers of potential therapeutic interest include lactoferrin, CXCR1, CXCR2, MMP-9, NGAL, elafin, HNE, pANCAs, MPO, CD16, CD177, CD64, HNPs, SLPI, and PTX3 [252,253,254].
In addition to the biomarker calprotectin, some other biomarkers have been related to inflammatory bowel disease. The serum levels of the peptide adropin, which acts as an energy regulator through lipid and glucose metabolism [255] and is considered an inflammatory biomarker [256], are reduced in patients with inflammatory bowel diseases, showing a negative correlation with fecal calprotectin [257]. Moreover, serum catestatin levels are increased in patients with inflammatory bowel disease when compared to control subjects [258,259,260]. Catestatin is a peptide proteolytically cleaved from chromogranin A, that primarily acts as an inhibitor of catecholamine secretion, and as stimulator of histamine release [261]. Enterochromaffin cells (EC) in the intestinal epithelium are a major source of chromogranin A [262]. Chromogranin A and catestatin regulate gut permeability via the antagonistic actions of its proteolytic peptides [259]. Catestatin regulates epithelial cell dynamics [263], and alters gut microbiota composition in mice [264]. Human catestatin also regulates intestinal inflammation via the macrophage population and through a STAT-3 dependent pathway in a murine model of colitis [260].
The mucosal immune system is the most extensive part of the immune system. Contrasting the situation in the systemic immune system, intestinal immune cells are involved in a highly balanced immune response aimed at controlling pathogen invasion, while stopping excessive immune responses against innocuous food antigens and commensal microbes that could risk unintentional tissue injury.
Some intestinal cell populations can adjust their functions to the needs of the intestinal microenvironment under steady state, and even modify their phenotype and behavior to adapt to inflammatory conditions. This adaptation can be harmful in IBD, but is also a potential therapeutic target for the treatment of the disease.
This is the case of intestinal macrophages, which restrain their robust pro-inflammatory potential through a natural resistance to producing inflammatory mediators in response to pattern-recognition molecules, while also retaining several of their homeostatic abilities, including scavenging and phagocytosing bacteria, preserving Tregs and maintaining tolerance, and promoting epithelial cell renewal [265]. In the intestinal microenvironment, macrophages adapt their functions to the context. For example, CX3CR1high macrophages can distinguish harmful from commensal bacteria via TLR and NLR recognition. In the intestinal microenvironment, CX3CR1high macrophages are excellent phagocytes, but produce low levels of pro-inflammatory cytokines and maintain tolerance through the production of anti-inflammatory cytokines such as IL-10 [111]. CX3CR1high intestinal macrophages sense and take up bacterial antigen from the intestinal lumen via their transepithelial dendrites [135,136,137,138,139,140]. In homeostasis, the intestinal microbiota inhibit the migration of antigen-loaded CX3CR1high intestinal macrophages to mesenteric lymph nodes, thereby also inhibiting antigen presentation to T cells, and effectively sustaining tolerance towards commensal bacteria.
If the intestinal microbiota is impaired or exposed to inflammatory conditions, CX3CR1high macrophages can differentiate to pro-inflammatory effector cells and acquire the capacity to migrate to lymph nodes and present antigens to lymphocytes, which are critically involved in the development of IBD [121]. However, it is not known whether resident macrophages modify their phenotype during inflammation according to the microenvironment [122,266], or if there are distinct macrophage populations that perform distinct functions [90]. The continuous replenishment of intestinal macrophages from monocytes might facilitate plasticity and adaptation of macrophage populations in response to signals from the local intestinal microenvironment, and this might modify macrophage properties such as migration, cytokine-release profile, antigen presentation, and T and B cell activation, and tissue healing. The use of scRNA-seq helps to define the contribution of several cell types to IBD, including macrophages and other innate immune cells. scRNA-seq studies provide precise knowledge of the spatial immune specialization and dysregulated immune response during IBD at single-cell resolution. For example, this technique has revealed that macrophages and CD8+ T cells in the lamina propria of the human colon during ulcerative colitis have an effector phenotype and are activated, while their lipid metabolism is suppressed compared with these cells in the epithelial layer [248]. Given the complexity of the mucosal environment and the peculiarity of its immune populations, tailored therapeutic strategies will require better knowledge of the networks regulating this delicate intestinal balance.
Biological treatments have provided an effective therapeutic advance for many IBD patients; nevertheless, one-third of patients do not respond to therapy (known as primary non-responders). In addition, a subset of patients who initially respond to anti-TNF drugs discontinue therapy because they lose their response (secondary non-responders) or develop intolerance [267,268]. Although these limitations can be explained by low drug levels in the target tissue or immune responses to the treatment, the complexity of IBD pathophysiology is likely to contribute to these treatment failures. Therefore, it is essential to gain a better understanding of the cellular and molecular basis of IBD in order to achieve complete mucosal healing and reversal of inflammation.

Author Contributions

Conceptualization, J.M.G.-G.; writing, figure design, and editing, A.S., B.H.-F., R.G.-B., H.S.-M. and J.M.G.-G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Instituto de Salud Carlos III (ISCIII) (PI20/00306) with co-funding from the European Regional Development Fund (ERDF) “A way to build Europe”. The CNIC is supported by the ISCIII, the Ministerio de Ciencia, Innovación y Universidades (MCNU), and the Pro CNIC Foundation. B.H.-F. and R.G.-B. by the UAM and the MCNU FPU program (FPU18/00895, FPU19/01774); A.S. by Universidad Francisco de Vitoria; and H.S-M. by the Comunidad de Madrid YEI program (PEJ-2020-TL/BMD-17604).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created in this study. All the data reported in this review were found in original articles cited in the text.

Acknowledgments

A.S. and B.H.-F. contributed equally to this manuscript. The authors thank S. Bartlett for English editing. Parts of the figures were drawn by using pictures from Servier Medical Art accessed on 25 October 2022. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/). Authors thank Servier Medical Art for the creation of a great repertoire of medical images, under a CC BY 3.0 Unported License.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

APCAntigen presenting cell
Card9Caspase recruitment domain 9
CCLChemokine (C-C motif) ligand
CCRC-C motif chemokine receptor
CDCrohn’s disease
CitH3Citrullination histone H3
CLACutaneous lymphocyte antigen
CPDCommon dendritic progenitor
CXCLChemokine (C-X-C motif) ligand
CX3CRChemokine (C-X3-C motif) receptor
DAMPsDamage-associated molecular patterns
DCsDendritic cells
DSSDextran sodium sulfate
ECMExtracellular matrix
Flt3LFms-like tyrosine kinase 3 ligand
Foxp3Forkhead Box P3
G-CSFGranulocyte colony-stimulating factor
GM-CSFGranulocyte-macrophage colony-stimulating factor
GWASGenome-wide association studies
IBDInflammatory bowel disease
IFNInterferon
IgImmunoglobulin
IICInnate immune cells
iNOSInducible nitric oxide synthetase
ILCsInnate lymphoid cells
ILInterleukin
IRFInterferon regulatory factor
MCPMonocyte chemoattractant protein
MHCIIMajor histocompatibility complex molecules class II
MIPMacrophage inflammatory proteins
MMPsMatrix metalloproteinases
MMTMyofibroblast transition
MPOMyeloperoxidase
mTORMammalian target of rapamycin
NKNatural killer
NETsNeutrophil extracellular traps
NOD2Nucleotide binding oligomerization domain containing 2
NLRNod-like receptors
PAD4Protein arginine deiminase 4
PAMPsPathogen-associated molecular patterns
pDCPlasmacytoid DCs
RARetinoic acid
PRRsPattern recognition receptors
RORγtRAR-related orphan receptor gamma t
SCFAsShort-chain fatty acids
scRNA-seqSingle cell RNA sequencing
SIPRαSignal-regulatory protein alpha
STATSignal transducer and activator of transcription
ThT-helper
TGFTransforming growth factor
TL1ATNF-like ligand 1 A
TLRsToll-like receptors
TNBS2,4,6-trinitrobenzene sulfonic acid
TNFTumor necrosis factor
TregT regulatory
Tr1T regulatory type 1
TWASTranscriptome-wide association studies
UCUlcerative colitis
WNTWingless-related integration site
XCR1Chemokine (C motif) receptor 1

References

  1. Ananthakrishnan, A.N.; Bernstein, C.N.; Iliopoulos, D.; Macpherson, A.; Neurath, M.F.; Ali, R.A.R.; Vavricka, S.R.; Fiocchi, C. Environmental triggers in IBD: A review of progress and evidence. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 39–49. [Google Scholar] [CrossRef] [PubMed]
  2. Levine, A.; Griffiths, A.; Markowitz, J.; Wilson, D.C.; Turner, D.; Russell, R.K.; Fell, J.; Ruemmele, F.M.; Walters, T.; Sherlock, M.; et al. Pediatric modification of the Montreal classification for inflammatory bowel disease: The Paris classification. Inflamm. Bowel Dis. 2011, 17, 1314–1321. [Google Scholar] [CrossRef] [PubMed]
  3. Louis, E.; Van Kemseke, C.; Reenaers, C. Necessity of phenotypic classification of inflammatory bowel disease. Best Pract. Res. Clin. Gastroenterol. 2011, 25 (Suppl. 1), S2–S7. [Google Scholar] [CrossRef] [PubMed]
  4. Satsangi, J.; Silverberg, M.S.; Vermeire, S.; Colombel, J.F. The Montreal classification of inflammatory bowel disease: Controversies, consensus, and implications. Gut 2006, 55, 749–753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Molodecky, N.A.; Soon, I.S.; Rabi, D.M.; Ghali, W.A.; Ferris, M.; Chernoff, G.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Barkema, H.W.; et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology 2012, 142, 46–54.e42, quiz e30. [Google Scholar] [CrossRef] [Green Version]
  6. Ananthakrishnan, A.N. Epidemiology and risk factors for IBD. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 205–217. [Google Scholar] [CrossRef]
  7. Chang, J.T. Pathophysiology of Inflammatory Bowel Diseases. N. Engl. J. Med. 2020, 383, 2652–2664. [Google Scholar] [CrossRef]
  8. Pullan, R.D.; Thomas, G.A.; Rhodes, M.; Newcombe, R.G.; Williams, G.T.; Allen, A.; Rhodes, J. Thickness of adherent mucus gel on colonic mucosa in humans and its relevance to colitis. Gut 1994, 35, 353–359. [Google Scholar] [CrossRef] [Green Version]
  9. Parikh, K.; Antanaviciute, A.; Fawkner-Corbett, D.; Jagielowicz, M.; Aulicino, A.; Lagerholm, C.; Davis, S.; Kinchen, J.; Chen, H.H.; Alham, N.K.; et al. Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature 2019, 567, 49–55. [Google Scholar] [CrossRef]
  10. Kinchen, J.; Chen, H.H.; Parikh, K.; Antanaviciute, A.; Jagielowicz, M.; Fawkner-Corbett, D.; Ashley, N.; Cubitt, L.; Mellado-Gomez, E.; Attar, M.; et al. Structural Remodeling of the Human Colonic Mesenchyme in Inflammatory Bowel Disease. Cell 2018, 175, 372–386.e17. [Google Scholar] [CrossRef]
  11. Gustafsson, J.K.; Johansson, M.E.V. The role of goblet cells and mucus in intestinal homeostasis. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 785–803. [Google Scholar] [CrossRef]
  12. Ghilas, S.; O’Keefe, R.; Mielke, L.A.; Raghu, D.; Buchert, M.; Ernst, M. Crosstalk between epithelium, myeloid and innate lymphoid cells during gut homeostasis and disease. Front. Immunol. 2022, 13, 944982. [Google Scholar] [CrossRef]
  13. Bassler, K.; Schulte-Schrepping, J.; Warnat-Herresthal, S.; Aschenbrenner, A.C.; Schultze, J.L. The Myeloid Cell Compartment-Cell by Cell. Annu. Rev. Immunol. 2019, 37, 269–293. [Google Scholar] [CrossRef]
  14. Herrero-Fernandez, B.; Gomez-Bris, R.; Somovilla-Crespo, B.; Gonzalez-Granado, J.M. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int. J. Mol. Sci. 2019, 20, 5293. [Google Scholar] [CrossRef] [Green Version]
  15. Leppkes, M.; Neurath, M.F. Cytokines in inflammatory bowel diseases—Update 2020. Pharmacol. Res. 2020, 158, 104835. [Google Scholar] [CrossRef]
  16. Mann, E.A.; Saeed, S.A. Gastrointestinal infection as a trigger for inflammatory bowel disease. Curr. Opin. Gastroenterol. 2012, 28, 24–29. [Google Scholar] [CrossRef]
  17. Schroeder, B.O.; Birchenough, G.M.H.; Stahlman, M.; Arike, L.; Johansson, M.E.V.; Hansson, G.C.; Backhed, F. Bifidobacteria or Fiber Protects against Diet-Induced Microbiota-Mediated Colonic Mucus Deterioration. Cell Host Microbe 2018, 23, 27–40.e7. [Google Scholar] [CrossRef] [Green Version]
  18. Neurath, M.F.; Leppkes, M. Resolution of ulcerative colitis. Semin. Immunopathol. 2019, 41, 747–756. [Google Scholar] [CrossRef]
  19. Wen, Z.; Fiocchi, C. Inflammatory bowel disease: Autoimmune or immune-mediated pathogenesis? Clin. Dev. Immunol. 2004, 11, 195–204. [Google Scholar] [CrossRef]
  20. Turner, J.R. Intestinal mucosal barrier function in health and disease. Nat. Rev. Immunol. 2009, 9, 799–809. [Google Scholar] [CrossRef]
  21. Munoz, L.E.; Leppkes, M.; Fuchs, T.A.; Hoffmann, M.; Herrmann, M. Missing in action-The meaning of cell death in tissue damage and inflammation. Immunol. Rev. 2017, 280, 26–40. [Google Scholar] [CrossRef] [PubMed]
  22. Zhou, G.; Yu, L.; Fang, L.; Yang, W.; Yu, T.; Miao, Y.; Chen, M.; Wu, K.; Chen, F.; Cong, Y.; et al. CD177(+) neutrophils as functionally activated neutrophils negatively regulate IBD. Gut 2018, 67, 1052–1063. [Google Scholar] [CrossRef] [PubMed]
  23. Park, H.; Li, Z.; Yang, X.O.; Chang, S.H.; Nurieva, R.; Wang, Y.H.; Wang, Y.; Hood, L.; Zhu, Z.; Tian, Q.; et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat. Immunol. 2005, 6, 1133–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mateer, S.W.; Mathe, A.; Bruce, J.; Liu, G.; Maltby, S.; Fricker, M.; Goggins, B.J.; Tay, H.L.; Marks, E.; Burns, G.; et al. IL-6 Drives Neutrophil-Mediated Pulmonary Inflammation Associated with Bacteremia in Murine Models of Colitis. Am. J. Pathol. 2018, 188, 1625–1639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Salem, M.; El Azreq, M.A.; Pelletier, J.; Robaye, B.; Aoudjit, F.; Sevigny, J. Exacerbated intestinal inflammation in P2Y6 deficient mice is associated with Th17 activation. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 2595–2605. [Google Scholar] [CrossRef]
  26. Kamp, M.E.; Shim, R.; Nicholls, A.J.; Oliveira, A.C.; Mason, L.J.; Binge, L.; Mackay, C.R.; Wong, C.H. G Protein-Coupled Receptor 43 Modulates Neutrophil Recruitment during Acute Inflammation. PLoS ONE 2016, 11, e0163750. [Google Scholar] [CrossRef] [Green Version]
  27. Correa, R.O.; Vieira, A.; Sernaglia, E.M.; Lancellotti, M.; Vieira, A.T.; Avila-Campos, M.J.; Rodrigues, H.G.; Vinolo, M.A.R. Bacterial short-chain fatty acid metabolites modulate the inflammatory response against infectious bacteria. Cell. Microbiol. 2017, 19, e12720. [Google Scholar] [CrossRef] [Green Version]
  28. Bedouhene, S.; Liu, M.; Senani, N.; Boussetta, T.; Pintard, C.; Dang, P.M.; El-Benna, J. Prolyl-Isomerase Pin1 Controls Key fMLP-Induced Neutrophil Functions. Biomedicines 2021, 9, 1130. [Google Scholar] [CrossRef]
  29. Yuen, J.; Pluthero, F.G.; Douda, D.N.; Riedl, M.; Cherry, A.; Ulanova, M.; Kahr, W.H.; Palaniyar, N.; Licht, C. NETosing Neutrophils Activate Complement Both on Their Own NETs and Bacteria via Alternative and Non-alternative Pathways. Front. Immunol. 2016, 7, 137. [Google Scholar] [CrossRef] [Green Version]
  30. Matoszka, N.; Dzialo, J.; Tokarz-Deptula, B.; Deptula, W. NET and NETosis—New phenomenon in immunology. Postepy. Hig. Med. Dosw. 2012, 66, 437–445. [Google Scholar] [CrossRef]
  31. Dinallo, V.; Marafini, I.; Di Fusco, D.; Laudisi, F.; Franze, E.; Di Grazia, A.; Figliuzzi, M.M.; Caprioli, F.; Stolfi, C.; Monteleone, I.; et al. Neutrophil Extracellular Traps Sustain Inflammatory Signals in Ulcerative Colitis. J. Crohn’s Colitis 2019, 13, 772–784. [Google Scholar] [CrossRef]
  32. Kaur, A.; Goggolidou, P. Ulcerative colitis: Understanding its cellular pathology could provide insights into novel therapies. J. Inflamm. 2020, 17, 15. [Google Scholar] [CrossRef] [Green Version]
  33. Scannell, M.; Flanagan, M.B.; de Stefani, A.; Wynne, K.J.; Cagney, G.; Godson, C.; Maderna, P. Annexin-1 and peptide derivatives are released by apoptotic cells and stimulate phagocytosis of apoptotic neutrophils by macrophages. J. Immunol. 2007, 178, 4595–4605. [Google Scholar] [CrossRef] [Green Version]
  34. Watanabe, S.; Alexander, M.; Misharin, A.V.; Budinger, G.R.S. The role of macrophages in the resolution of inflammation. J. Clin. Investig. 2019, 129, 2619–2628. [Google Scholar] [CrossRef] [Green Version]
  35. McCracken, J.M.; Allen, L.A. Regulation of human neutrophil apoptosis and lifespan in health and disease. J. Cell Death 2014, 7, 15–23. [Google Scholar] [CrossRef] [Green Version]
  36. Urban, C.F.; Ermert, D.; Schmid, M.; Abu-Abed, U.; Goosmann, C.; Nacken, W.; Brinkmann, V.; Jungblut, P.R.; Zychlinsky, A. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009, 5, e1000639. [Google Scholar] [CrossRef] [Green Version]
  37. Chen, F.; Liu, Y.; Shi, Y.; Zhang, J.; Liu, X.; Liu, Z.; Lv, J.; Leng, Y. The emerging role of neutrophilic extracellular traps in intestinal disease. Gut Pathog. 2022, 14, 27. [Google Scholar] [CrossRef]
  38. Jorch, S.K.; Kubes, P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat. Med. 2017, 23, 279–287. [Google Scholar] [CrossRef]
  39. Drury, B.; Hardisty, G.; Gray, R.D.; Ho, G.T. Neutrophil Extracellular Traps in Inflammatory Bowel Disease: Pathogenic Mechanisms and Clinical Translation. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 321–333. [Google Scholar] [CrossRef]
  40. Bennike, T.B.; Carlsen, T.G.; Ellingsen, T.; Bonderup, O.K.; Glerup, H.; Bogsted, M.; Christiansen, G.; Birkelund, S.; Stensballe, A.; Andersen, V. Neutrophil Extracellular Traps in Ulcerative Colitis: A Proteome Analysis of Intestinal Biopsies. Inflamm. Bowel Dis. 2015, 21, 2052–2067. [Google Scholar] [CrossRef]
  41. Maronek, M.; Gromova, B.; Liptak, R.; Konecna, B.; Pastorek, M.; Cechova, B.; Harsanyova, M.; Budis, J.; Smolak, D.; Radvanszky, J.; et al. Extracellular DNA Correlates with Intestinal Inflammation in Chemically Induced Colitis in Mice. Cells 2021, 10, 81. [Google Scholar] [CrossRef] [PubMed]
  42. Hansberry, D.R.; Shah, K.; Agarwal, P.; Agarwal, N. Fecal Myeloperoxidase as a Biomarker for Inflammatory Bowel Disease. Cureus 2017, 9, e1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Abd El Hafez, A.; Mohamed, A.S.; Shehta, A.; Sheta, H. Neutrophil extracellular traps-associated protein peptidyl arginine deaminase 4 immunohistochemical expression in ulcerative colitis and its association with the prognostic predictors. Pathol. Res. Pract. 2020, 216, 153102. [Google Scholar] [CrossRef] [PubMed]
  44. Kaluzna, A.; Olczyk, P.; Komosinska-Vassev, K. The Role of Innate and Adaptive Immune Cells in the Pathogenesis and Development of the Inflammatory Response in Ulcerative Colitis. J. Clin. Med. 2022, 11, 400. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, T.; Mei, Y.; Dong, W.; Wang, J.; Huang, F.; Wu, J. Evaluation of protein arginine deiminase-4 inhibitor in TNBS- induced colitis in mice. Int. Immunopharmacol. 2020, 84, 106583. [Google Scholar] [CrossRef] [PubMed]
  46. Li, T.; Wang, C.; Liu, Y.; Li, B.; Zhang, W.; Wang, L.; Yu, M.; Zhao, X.; Du, J.; Zhang, J.; et al. Neutrophil Extracellular Traps Induce Intestinal Damage and Thrombotic Tendency in Inflammatory Bowel Disease. J. Crohn’s Colitis 2020, 14, 240–253. [Google Scholar] [CrossRef]
  47. Lin, E.Y.; Lai, H.J.; Cheng, Y.K.; Leong, K.Q.; Cheng, L.C.; Chou, Y.C.; Peng, Y.C.; Hsu, Y.H.; Chiang, H.S. Neutrophil Extracellular Traps Impair Intestinal Barrier Function during Experimental Colitis. Biomedicines 2020, 8, 275. [Google Scholar] [CrossRef]
  48. Dong, W.; Liu, D.; Zhang, T.; You, Q.; Huang, F.; Wu, J. Oral delivery of staphylococcal nuclease ameliorates DSS induced ulcerative colitis in mice via degrading intestinal neutrophil extracellular traps. Ecotoxicol. Environ. Saf. 2021, 215, 112161. [Google Scholar] [CrossRef]
  49. Li, G.; Lin, J.; Zhang, C.; Gao, H.; Lu, H.; Gao, X.; Zhu, R.; Li, Z.; Li, M.; Liu, Z. Microbiota metabolite butyrate constrains neutrophil functions and ameliorates mucosal inflammation in inflammatory bowel disease. Gut Microbes 2021, 13, 1968257. [Google Scholar] [CrossRef]
  50. Biasi, F.; Leonarduzzi, G.; Oteiza, P.I.; Poli, G. Inflammatory bowel disease: Mechanisms, redox considerations, and therapeutic targets. Antioxid. Redox Signal. 2013, 19, 1711–1747. [Google Scholar] [CrossRef]
  51. Wera, O.; Lancellotti, P.; Oury, C. The Dual Role of Neutrophils in Inflammatory Bowel Diseases. J. Clin. Med. 2016, 5, 118. [Google Scholar] [CrossRef]
  52. Zhou, G.X.; Liu, Z.J. Potential roles of neutrophils in regulating intestinal mucosal inflammation of inflammatory bowel disease. J. Dig. Dis. 2017, 18, 495–503. [Google Scholar] [CrossRef] [Green Version]
  53. Pavlidis, P.; Tsakmaki, A.; Pantazi, E.; Li, K.; Cozzetto, D.; Digby-Bell, J.; Yang, F.; Lo, J.W.; Alberts, E.; Sa, A.C.C.; et al. Interleukin-22 regulates neutrophil recruitment in ulcerative colitis and is associated with resistance to ustekinumab therapy. Nat. Commun. 2022, 13, 5820. [Google Scholar] [CrossRef]
  54. Fenini, G.; Contassot, E.; French, L.E. Potential of IL-1, IL-18 and Inflammasome Inhibition for the Treatment of Inflammatory Skin Diseases. Front. Pharmacol. 2017, 8, 278. [Google Scholar] [CrossRef] [Green Version]
  55. Mao, L.; Kitani, A.; Strober, W.; Fuss, I.J. The Role of NLRP3 and IL-1beta in the Pathogenesis of Inflammatory Bowel Disease. Front. Immunol. 2018, 9, 2566. [Google Scholar] [CrossRef] [Green Version]
  56. Elinav, E.; Strowig, T.; Kau, A.L.; Henao-Mejia, J.; Thaiss, C.A.; Booth, C.J.; Peaper, D.R.; Bertin, J.; Eisenbarth, S.C.; Gordon, J.I.; et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 2011, 145, 745–757. [Google Scholar] [CrossRef] [Green Version]
  57. Di Paolo, N.C.; Shayakhmetov, D.M. Interleukin 1alpha and the inflammatory process. Nat. Immunol. 2016, 17, 906–913. [Google Scholar] [CrossRef] [Green Version]
  58. Laan, M.; Prause, O.; Miyamoto, M.; Sjostrand, M.; Hytonen, A.M.; Kaneko, T.; Lotvall, J.; Linden, A. A role of GM-CSF in the accumulation of neutrophils in the airways caused by IL-17 and TNF-alpha. Eur. Respir. J. 2003, 21, 387–393. [Google Scholar] [CrossRef] [Green Version]
  59. Leppkes, M.; Becker, C.; Ivanov, I.I.; Hirth, S.; Wirtz, S.; Neufert, C.; Pouly, S.; Murphy, A.J.; Valenzuela, D.M.; Yancopoulos, G.D.; et al. RORgamma-expressing Th17 cells induce murine chronic intestinal inflammation via redundant effects of IL-17A and IL-17F. Gastroenterology 2009, 136, 257–267. [Google Scholar] [CrossRef]
  60. Stark, M.A.; Huo, Y.; Burcin, T.L.; Morris, M.A.; Olson, T.S.; Ley, K. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity 2005, 22, 285–294. [Google Scholar] [CrossRef]
  61. Phillipson, M.; Kubes, P. The Healing Power of Neutrophils. Trends. Immunol. 2019, 40, 635–647. [Google Scholar] [CrossRef] [PubMed]
  62. Danne, C.; Michaudel, C.; Skerniskyte, J.; Planchais, J.; Magniez, A.; Agus, A.; Michel, M.L.; Lamas, B.; Da Costa, G.; Spatz, M.; et al. CARD9 in neutrophils protects from colitis and controls mitochondrial metabolism and cell survival. Gut 2022. [Google Scholar] [CrossRef] [PubMed]
  63. Kang, L.; Fang, X.; Song, Y.H.; He, Z.X.; Wang, Z.J.; Wang, S.L.; Li, Z.S.; Bai, Y. Neutrophil-Epithelial Crosstalk During Intestinal Inflammation. Cell. Mol. Gastroenterol. Hepatol. 2022, 14, 1257–1267. [Google Scholar] [CrossRef] [PubMed]
  64. McCourt, M.; Wang, J.H.; Sookhai, S.; Redmond, H.P. Proinflammatory mediators stimulate neutrophil-directed angiogenesis. Arch. Surg. 1999, 134, 1325–1331, discussion 1331–1332. [Google Scholar] [CrossRef] [PubMed]
  65. Schwab, J.M.; Chiang, N.; Arita, M.; Serhan, C.N. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature 2007, 447, 869–874. [Google Scholar] [CrossRef] [Green Version]
  66. Seo, D.H.; Che, X.; Kim, S.; Kim, D.H.; Ma, H.W.; Kim, J.H.; Kim, T.I.; Kim, W.H.; Kim, S.W.; Cheon, J.H. Triggering Receptor Expressed on Myeloid Cells-1 Agonist Regulates Intestinal Inflammation via Cd177(+) Neutrophils. Front. Immunol. 2021, 12, 650864. [Google Scholar] [CrossRef]
  67. Clancy, D.M.; Sullivan, G.P.; Moran, H.B.T.; Henry, C.M.; Reeves, E.P.; McElvaney, N.G.; Lavelle, E.C.; Martin, S.J. Extracellular Neutrophil Proteases are Efficient Regulators of IL-1, IL-33, and IL-36 Cytokine Activity but Poor Effectors of Microbial Killing. Cell Rep. 2018, 22, 2937–2950. [Google Scholar] [CrossRef] [Green Version]
  68. Yang, D.; Chen, Q.; Chertov, O.; Oppenheim, J.J. Human neutrophil defensins selectively chemoattract naive T and immature dendritic cells. J. Leukoc. Biol. 2000, 68, 9–14. [Google Scholar] [CrossRef]
  69. Cunliffe, R.N.; Kamal, M.; Rose, F.R.; James, P.D.; Mahida, Y.R. Expression of antimicrobial neutrophil defensins in epithelial cells of active inflammatory bowel disease mucosa. J. Clin. Pathol. 2002, 55, 298–304. [Google Scholar] [CrossRef]
  70. Yui, S.; Nakatani, Y.; Mikami, M. Calprotectin (S100A8/S100A9), an inflammatory protein complex from neutrophils with a broad apoptosis-inducing activity. Biol. Pharm. Bull. 2003, 26, 753–760. [Google Scholar] [CrossRef]
  71. Karmakar, M.; Minns, M.; Greenberg, E.N.; Diaz-Aponte, J.; Pestonjamasp, K.; Johnson, J.L.; Rathkey, J.K.; Abbott, D.W.; Wang, K.; Shao, F.; et al. N-GSDMD trafficking to neutrophil organelles facilitates IL-1beta release independently of plasma membrane pores and pyroptosis. Nat. Commun. 2020, 11, 2212. [Google Scholar] [CrossRef]
  72. Ozaki, R.; Kobayashi, T.; Okabayashi, S.; Nakano, M.; Morinaga, S.; Hara, A.; Ohbu, M.; Matsuoka, K.; Toyonaga, T.; Saito, E.; et al. Histological Risk Factors to Predict Clinical Relapse in Ulcerative Colitis with Endoscopically Normal Mucosa. J. Crohn’s Colitis 2018, 12, 1288–1294. [Google Scholar] [CrossRef]
  73. Surawicz, C.M.; Haggitt, R.C.; Husseman, M.; McFarland, L.V. Mucosal biopsy diagnosis of colitis: Acute self-limited colitis and idiopathic inflammatory bowel disease. Gastroenterology 1994, 107, 755–763. [Google Scholar] [CrossRef]
  74. Shinoda, M.; Shin-Ya, M.; Naito, Y.; Kishida, T.; Ito, R.; Suzuki, N.; Yasuda, H.; Sakagami, J.; Imanishi, J.; Kataoka, K.; et al. Early-stage blocking of Notch signaling inhibits the depletion of goblet cells in dextran sodium sulfate-induced colitis in mice. J. Gastroenterol. 2010, 45, 608–617. [Google Scholar] [CrossRef]
  75. Leiper, K.; Campbell, B.J.; Jenkinson, M.D.; Milton, J.; Yu, L.G.; Democratis, J.; Rhodes, J.M. Interaction between bacterial peptides, neutrophils and goblet cells: A possible mechanism for neutrophil recruitment and goblet cell depletion in colitis. Clin. Sci. 2001, 101, 395–402. [Google Scholar] [CrossRef]
  76. Bilyy, R.; Fedorov, V.; Vovk, V.; Leppkes, M.; Dumych, T.; Chopyak, V.; Schett, G.; Herrmann, M. Neutrophil Extracellular Traps Form a Barrier between Necrotic and Viable Areas in Acute Abdominal Inflammation. Front. Immunol. 2016, 7, 424. [Google Scholar] [CrossRef] [Green Version]
  77. Bain, C.C.; Schridde, A. Origin, Differentiation, and Function of Intestinal Macrophages. Front. Immunol. 2018, 9, 2733. [Google Scholar] [CrossRef] [Green Version]
  78. Bleriot, C.; Chakarov, S.; Ginhoux, F. Determinants of Resident Tissue Macrophage Identity and Function. Immunity 2020, 52, 957–970. [Google Scholar] [CrossRef]
  79. Lis-Lopez, L.; Bauset, C.; Seco-Cervera, M.; Cosin-Roger, J. Is the Macrophage Phenotype Determinant for Fibrosis Development? Biomedicines 2021, 9, 1747. [Google Scholar] [CrossRef]
  80. Gomez Perdiguero, E.; Klapproth, K.; Schulz, C.; Busch, K.; Azzoni, E.; Crozet, L.; Garner, H.; Trouillet, C.; de Bruijn, M.F.; Geissmann, F.; et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 2015, 518, 547–551. [Google Scholar] [CrossRef]
  81. Guilliams, M.; Scott, C.L. Does niche competition determine the origin of tissue-resident macrophages? Nat. Rev. Immunol. 2017, 17, 451–460. [Google Scholar] [CrossRef] [PubMed]
  82. Epelman, S.; Lavine, K.J.; Randolph, G.J. Origin and functions of tissue macrophages. Immunity 2014, 41, 21–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Haldar, M.; Murphy, K.M. Origin, development, and homeostasis of tissue-resident macrophages. Immunol. Rev. 2014, 262, 25–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef] [PubMed]
  85. Locati, M.; Curtale, G.; Mantovani, A. Diversity, Mechanisms, and Significance of Macrophage Plasticity. Annu. Rev. Pathol. 2020, 15, 123–147. [Google Scholar] [CrossRef] [Green Version]
  86. Perdiguero, E.G.; Geissmann, F. The development and maintenance of resident macrophages. Nat. Immunol. 2016, 17, 2–8. [Google Scholar] [CrossRef] [Green Version]
  87. Ginhoux, F.; Guilliams, M. Tissue-Resident Macrophage Ontogeny and Homeostasis. Immunity 2016, 44, 439–449. [Google Scholar] [CrossRef]
  88. De Schepper, S.; Verheijden, S.; Aguilera-Lizarraga, J.; Viola, M.F.; Boesmans, W.; Stakenborg, N.; Voytyuk, I.; Schmidt, I.; Boeckx, B.; Dierckx de Casterle, I.; et al. Self-Maintaining Gut Macrophages Are Essential for Intestinal Homeostasis. Cell 2018, 175, 400–415.e13. [Google Scholar] [CrossRef] [Green Version]
  89. Bain, C.C.; Bravo-Blas, A.; Scott, C.L.; Perdiguero, E.G.; Geissmann, F.; Henri, S.; Malissen, B.; Osborne, L.C.; Artis, D.; Mowat, A.M. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nat. Immunol. 2014, 15, 929–937. [Google Scholar] [CrossRef] [Green Version]
  90. Bain, C.C.; Scott, C.L.; Uronen-Hansson, H.; Gudjonsson, S.; Jansson, O.; Grip, O.; Guilliams, M.; Malissen, B.; Agace, W.W.; Mowat, A.M. Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal Immunol. 2013, 6, 498–510. [Google Scholar] [CrossRef]
  91. Tamoutounour, S.; Henri, S.; Lelouard, H.; de Bovis, B.; de Haar, C.; van der Woude, C.J.; Woltman, A.M.; Reyal, Y.; Bonnet, D.; Sichien, D.; et al. CD64 distinguishes macrophages from dendritic cells in the gut and reveals the Th1-inducing role of mesenteric lymph node macrophages during colitis. Eur. J. Immunol. 2012, 42, 3150–3166. [Google Scholar] [CrossRef]
  92. Delfini, M.; Stakenborg, N.; Viola, M.F.; Boeckxstaens, G. Macrophages in the gut: Masters in multitasking. Immunity 2022, 55, 1530–1548. [Google Scholar] [CrossRef]
  93. Viola, M.F.; Boeckxstaens, G. Niche-specific functional heterogeneity of intestinal resident macrophages. Gut 2021, 70, 1383–1395. [Google Scholar] [CrossRef]
  94. Gabanyi, I.; Muller, P.A.; Feighery, L.; Oliveira, T.Y.; Costa-Pinto, F.A.; Mucida, D. Neuro-immune Interactions Drive Tissue Programming in Intestinal Macrophages. Cell 2016, 164, 378–391. [Google Scholar] [CrossRef] [Green Version]
  95. Na, Y.R.; Stakenborg, M.; Seok, S.H.; Matteoli, G. Macrophages in intestinal inflammation and resolution: A potential therapeutic target in IBD. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 531–543. [Google Scholar] [CrossRef]
  96. Shaw, T.N.; Houston, S.A.; Wemyss, K.; Bridgeman, H.M.; Barbera, T.A.; Zangerle-Murray, T.; Strangward, P.; Ridley, A.J.L.; Wang, P.; Tamoutounour, S.; et al. Tissue-resident macrophages in the intestine are long lived and defined by Tim-4 and CD4 expression. J. Exp. Med. 2018, 215, 1507–1518. [Google Scholar] [CrossRef]
  97. Muller, P.A.; Koscso, B.; Rajani, G.M.; Stevanovic, K.; Berres, M.L.; Hashimoto, D.; Mortha, A.; Leboeuf, M.; Li, X.M.; Mucida, D.; et al. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cell 2014, 158, 300–313. [Google Scholar] [CrossRef] [Green Version]
  98. Hume, D.A.; Perry, V.H.; Gordon, S. The mononuclear phagocyte system of the mouse defined by immunohistochemical localisation of antigen F4/80: Macrophages associated with epithelia. Anat. Rec. 1984, 210, 503–512. [Google Scholar] [CrossRef]
  99. Hayashi, A.; Sato, T.; Kamada, N.; Mikami, Y.; Matsuoka, K.; Hisamatsu, T.; Hibi, T.; Roers, A.; Yagita, H.; Ohteki, T.; et al. A single strain of Clostridium butyricum induces intestinal IL-10-producing macrophages to suppress acute experimental colitis in mice. Cell Host Microbe 2013, 13, 711–722. [Google Scholar] [CrossRef] [Green Version]
  100. Danne, C.; Ryzhakov, G.; Martinez-Lopez, M.; Ilott, N.E.; Franchini, F.; Cuskin, F.; Lowe, E.C.; Bullers, S.J.; Arthur, J.S.C.; Powrie, F. A Large Polysaccharide Produced by Helicobacter hepaticus Induces an Anti-inflammatory Gene Signature in Macrophages. Cell Host Microbe 2017, 22, 733–745.e5. [Google Scholar] [CrossRef]
  101. Hiemstra, I.H.; Beijer, M.R.; Veninga, H.; Vrijland, K.; Borg, E.G.; Olivier, B.J.; Mebius, R.E.; Kraal, G.; den Haan, J.M. The identification and developmental requirements of colonic CD169(+) macrophages. Immunology 2014, 142, 269–278. [Google Scholar] [CrossRef] [PubMed]
  102. Sehgal, A.; Donaldson, D.S.; Pridans, C.; Sauter, K.A.; Hume, D.A.; Mabbott, N.A. The role of CSF1R-dependent macrophages in control of the intestinal stem-cell niche. Nat. Commun. 2018, 9, 1272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Hadis, U.; Wahl, B.; Schulz, O.; Hardtke-Wolenski, M.; Schippers, A.; Wagner, N.; Muller, W.; Sparwasser, T.; Forster, R.; Pabst, O. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity 2011, 34, 237–246. [Google Scholar] [CrossRef] [PubMed]
  104. Shaw, M.H.; Kamada, N.; Kim, Y.G.; Nunez, G. Microbiota-induced IL-1beta, but not IL-6, is critical for the development of steady-state TH17 cells in the intestine. J. Exp. Med. 2012, 209, 251–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. He, X.; Tan, S.; Shao, Z.; Wang, X. Latitudinal and longitudinal regulation of tissue macrophages in inflammatory diseases. Genes Dis. 2022, 9, 1194–1207. [Google Scholar] [CrossRef]
  106. Yao, H.; Tang, G. Macrophages in intestinal fibrosis and regression. Cell Immunol. 2022, 381, 104614. [Google Scholar] [CrossRef]
  107. Mortha, A.; Chudnovskiy, A.; Hashimoto, D.; Bogunovic, M.; Spencer, S.P.; Belkaid, Y.; Merad, M. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 2014, 343, 1249288. [Google Scholar] [CrossRef] [Green Version]
  108. Spindler, M.P.; Siu, S.; Mogno, I.; Li, Z.; Yang, C.; Mehandru, S.; Britton, G.J.; Faith, J.J. Human gut microbiota stimulate defined innate immune responses that vary from phylum to strain. Cell Host Microbe 2022, 30, 1481–1498.e5. [Google Scholar] [CrossRef]
  109. Zegarra Ruiz, D.F.; Kim, D.V.; Norwood, K.; Saldana-Morales, F.B.; Kim, M.; Ng, C.; Callaghan, R.; Uddin, M.; Chang, L.C.; Longman, R.S.; et al. Microbiota manipulation to increase macrophage IL-10 improves colitis and limits colitis-associated colorectal cancer. Gut Microbes 2022, 14, 2119054. [Google Scholar] [CrossRef]
  110. Mosser, D.M.; Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 2008, 8, 958–969. [Google Scholar] [CrossRef]
  111. Ruder, B.; Becker, C. At the Forefront of the Mucosal Barrier: The Role of Macrophages in the Intestine. Cells 2020, 9, 2162. [Google Scholar] [CrossRef]
  112. Miao, E.A.; Mao, D.P.; Yudkovsky, N.; Bonneau, R.; Lorang, C.G.; Warren, S.E.; Leaf, I.A.; Aderem, A. Innate immune detection of the type III secretion apparatus through the NLRC4 inflammasome. Proc. Natl. Acad. Sci. USA 2010, 107, 3076–3080. [Google Scholar] [CrossRef] [Green Version]
  113. Bortoluci, K.R.; Medzhitov, R. Control of infection by pyroptosis and autophagy: Role of TLR and NLR. Cell Mol. Life Sci. 2010, 67, 1643–1651. [Google Scholar] [CrossRef]
  114. Rivollier, A.; He, J.; Kole, A.; Valatas, V.; Kelsall, B.L. Inflammation switches the differentiation program of Ly6Chi monocytes from antiinflammatory macrophages to inflammatory dendritic cells in the colon. J. Exp. Med. 2012, 209, 139–155. [Google Scholar] [CrossRef] [Green Version]
  115. Smith, P.D.; Smythies, L.E.; Mosteller-Barnum, M.; Sibley, D.A.; Russell, M.W.; Merger, M.; Sellers, M.T.; Orenstein, J.M.; Shimada, T.; Graham, M.F.; et al. Intestinal macrophages lack CD14 and CD89 and consequently are down-regulated for LPS- and IgA-mediated activities. J. Immunol. 2001, 167, 2651–2656. [Google Scholar] [CrossRef] [Green Version]
  116. Weber, B.; Saurer, L.; Schenk, M.; Dickgreber, N.; Mueller, C. CX3CR1 defines functionally distinct intestinal mononuclear phagocyte subsets which maintain their respective functions during homeostatic and inflammatory conditions. Eur. J. Immunol. 2011, 41, 773–779. [Google Scholar] [CrossRef]
  117. Smythies, L.E.; Sellers, M.; Clements, R.H.; Mosteller-Barnum, M.; Meng, G.; Benjamin, W.H.; Orenstein, J.M.; Smith, P.D. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J. Clin. Investig. 2005, 115, 66–75. [Google Scholar] [CrossRef] [Green Version]
  118. Denning, T.L.; Wang, Y.C.; Patel, S.R.; Williams, I.R.; Pulendran, B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat. Immunol. 2007, 8, 1086–1094. [Google Scholar] [CrossRef]
  119. Murai, M.; Turovskaya, O.; Kim, G.; Madan, R.; Karp, C.L.; Cheroutre, H.; Kronenberg, M. Interleukin 10 acts on regulatory T cells to maintain expression of the transcription factor Foxp3 and suppressive function in mice with colitis. Nat. Immunol. 2009, 10, 1178–1184. [Google Scholar] [CrossRef] [Green Version]
  120. Ueda, Y.; Kayama, H.; Jeon, S.G.; Kusu, T.; Isaka, Y.; Rakugi, H.; Yamamoto, M.; Takeda, K. Commensal microbiota induce LPS hyporesponsiveness in colonic macrophages via the production of IL-10. Int. Immunol. 2010, 22, 953–962. [Google Scholar] [CrossRef]
  121. Zigmond, E.; Bernshtein, B.; Friedlander, G.; Walker, C.R.; Yona, S.; Kim, K.W.; Brenner, O.; Krauthgamer, R.; Varol, C.; Muller, W.; et al. Macrophage-restricted interleukin-10 receptor deficiency, but not IL-10 deficiency, causes severe spontaneous colitis. Immunity 2014, 40, 720–733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Zigmond, E.; Varol, C.; Farache, J.; Elmaliah, E.; Satpathy, A.T.; Friedlander, G.; Mack, M.; Shpigel, N.; Boneca, I.G.; Murphy, K.M.; et al. Ly6C hi monocytes in the inflamed colon give rise to proinflammatory effector cells and migratory antigen-presenting cells. Immunity 2012, 37, 1076–1090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Zhou, Z.; Ding, M.; Huang, L.; Gilkeson, G.; Lang, R.; Jiang, W. Toll-like receptor-mediated immune responses in intestinal macrophages; implications for mucosal immunity and autoimmune diseases. Clin. Immunol. 2016, 173, 81–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Lin, W.; Xu, D.; Austin, C.D.; Caplazi, P.; Senger, K.; Sun, Y.; Jeet, S.; Young, J.; Delarosa, D.; Suto, E.; et al. Function of CSF1 and IL34 in Macrophage Homeostasis, Inflammation, and Cancer. Front. Immunol. 2019, 10, 2019. [Google Scholar] [CrossRef] [PubMed]
  125. Zwicker, S.; Martinez, G.L.; Bosma, M.; Gerling, M.; Clark, R.; Majster, M.; Soderman, J.; Almer, S.; Bostrom, E.A. Interleukin 34: A new modulator of human and experimental inflammatory bowel disease. Clin. Sci. 2015, 129, 281–290. [Google Scholar] [CrossRef] [Green Version]
  126. Dai, X.M.; Ryan, G.R.; Hapel, A.J.; Dominguez, M.G.; Russell, R.G.; Kapp, S.; Sylvestre, V.; Stanley, E.R. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood 2002, 99, 111–120. [Google Scholar] [CrossRef]
  127. Felix, R.; Cecchini, M.G.; Hofstetter, W.; Elford, P.R.; Stutzer, A.; Fleisch, H. Impairment of macrophage colony-stimulating factor production and lack of resident bone marrow macrophages in the osteopetrotic op/op mouse. J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 1990, 5, 781–789. [Google Scholar] [CrossRef]
  128. Wiktor-Jedrzejczak, W.; Bartocci, A.; Ferrante, A.W., Jr.; Ahmed-Ansari, A.; Sell, K.W.; Pollard, J.W.; Stanley, E.R. Total absence of colony-stimulating factor 1 in the macrophage-deficient osteopetrotic (op/op) mouse. Proc. Natl. Acad. Sci. USA 1990, 87, 4828–4832. [Google Scholar] [CrossRef] [Green Version]
  129. Cecchini, M.G.; Dominguez, M.G.; Mocci, S.; Wetterwald, A.; Felix, R.; Fleisch, H.; Chisholm, O.; Hofstetter, W.; Pollard, J.W.; Stanley, E.R. Role of colony stimulating factor-1 in the establishment and regulation of tissue macrophages during postnatal development of the mouse. Development 1994, 120, 1357–1372. [Google Scholar] [CrossRef]
  130. MacDonald, K.P.; Palmer, J.S.; Cronau, S.; Seppanen, E.; Olver, S.; Raffelt, N.C.; Kuns, R.; Pettit, A.R.; Clouston, A.; Wainwright, B.; et al. An antibody against the colony-stimulating factor 1 receptor depletes the resident subset of monocytes and tissue- and tumor-associated macrophages but does not inhibit inflammation. Blood 2010, 116, 3955–3963. [Google Scholar] [CrossRef]
  131. Hume, D.A.; Pavli, P.; Donahue, R.E.; Fidler, I.J. The effect of human recombinant macrophage colony-stimulating factor (CSF-1) on the murine mononuclear phagocyte system in vivo. J. Immunol. 1988, 141, 3405–3409. [Google Scholar] [CrossRef]
  132. Schridde, A.; Bain, C.C.; Mayer, J.U.; Montgomery, J.; Pollet, E.; Denecke, B.; Milling, S.W.F.; Jenkins, S.J.; Dalod, M.; Henri, S.; et al. Tissue-specific differentiation of colonic macrophages requires TGFbeta receptor-mediated signaling. Mucosal Immunol. 2017, 10, 1387–1399. [Google Scholar] [CrossRef] [Green Version]
  133. Smith, P.D.; Janoff, E.N.; Mosteller-Barnum, M.; Merger, M.; Orenstein, J.M.; Kearney, J.F.; Graham, M.F. Isolation and purification of CD14-negative mucosal macrophages from normal human small intestine. J. Immunol. Methods 1997, 202, 1–11. [Google Scholar] [CrossRef]
  134. Cummings, R.J.; Barbet, G.; Bongers, G.; Hartmann, B.M.; Gettler, K.; Muniz, L.; Furtado, G.C.; Cho, J.; Lira, S.A.; Blander, J.M. Different tissue phagocytes sample apoptotic cells to direct distinct homeostasis programs. Nature 2016, 539, 565–569. [Google Scholar] [CrossRef]
  135. Vallon-Eberhard, A.; Landsman, L.; Yogev, N.; Verrier, B.; Jung, S. Transepithelial pathogen uptake into the small intestinal lamina propria. J. Immunol. 2006, 176, 2465–2469. [Google Scholar] [CrossRef] [Green Version]
  136. Rescigno, M.; Urbano, M.; Valzasina, B.; Francolini, M.; Rotta, G.; Bonasio, R.; Granucci, F.; Kraehenbuhl, J.P.; Ricciardi-Castagnoli, P. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat. Immunol. 2001, 2, 361–367. [Google Scholar] [CrossRef]
  137. Hapfelmeier, S.; Muller, A.J.; Stecher, B.; Kaiser, P.; Barthel, M.; Endt, K.; Eberhard, M.; Robbiani, R.; Jacobi, C.A.; Heikenwalder, M.; et al. Microbe sampling by mucosal dendritic cells is a discrete, MyD88-independent step in DeltainvG S. Typhimurium colitis. J. Exp. Med. 2008, 205, 437–450. [Google Scholar] [CrossRef] [Green Version]
  138. Medina-Contreras, O.; Geem, D.; Laur, O.; Williams, I.R.; Lira, S.A.; Nusrat, A.; Parkos, C.A.; Denning, T.L. CX3CR1 regulates intestinal macrophage homeostasis, bacterial translocation, and colitogenic Th17 responses in mice. J. Clin. Investig. 2011, 121, 4787–4795. [Google Scholar] [CrossRef] [Green Version]
  139. Pabst, O.; Bernhardt, G. The puzzle of intestinal lamina propria dendritic cells and macrophages. Eur. J. Immunol. 2010, 40, 2107–2111. [Google Scholar] [CrossRef]
  140. Niess, J.H.; Brand, S.; Gu, X.; Landsman, L.; Jung, S.; McCormick, B.A.; Vyas, J.M.; Boes, M.; Ploegh, H.L.; Fox, J.G.; et al. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 2005, 307, 254–258. [Google Scholar] [CrossRef]
  141. Mazzini, E.; Massimiliano, L.; Penna, G.; Rescigno, M. Oral tolerance can be established via gap junction transfer of fed antigens from CX3CR1(+) macrophages to CD103(+) dendritic cells. Immunity 2014, 40, 248–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Farache, J.; Koren, I.; Milo, I.; Gurevich, I.; Kim, K.W.; Zigmond, E.; Furtado, G.C.; Lira, S.A.; Shakhar, G. Luminal bacteria recruit CD103+ dendritic cells into the intestinal epithelium to sample bacterial antigens for presentation. Immunity 2013, 38, 581–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Denning, T.L.; Norris, B.A.; Medina-Contreras, O.; Manicassamy, S.; Geem, D.; Madan, R.; Karp, C.L.; Pulendran, B. Functional specializations of intestinal dendritic cell and macrophage subsets that control Th17 and regulatory T cell responses are dependent on the T cell/APC ratio, source of mouse strain, and regional localization. J. Immunol. 2011, 187, 733–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Panea, C.; Farkas, A.M.; Goto, Y.; Abdollahi-Roodsaz, S.; Lee, C.; Koscso, B.; Gowda, K.; Hohl, T.M.; Bogunovic, M.; Ivanov, I.I. Intestinal Monocyte-Derived Macrophages Control Commensal-Specific Th17 Responses. Cell. Rep. 2015, 12, 1314–1324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Serbina, N.V.; Pamer, E.G. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat. Immunol. 2006, 7, 311–317. [Google Scholar] [CrossRef]
  146. El Sayed, S.; Patik, I.; Redhu, N.S.; Glickman, J.N.; Karagiannis, K.; El Naenaeey, E.S.Y.; Elmowalid, G.A.; Abd El Wahab, A.M.; Snapper, S.B.; Horwitz, B.H. CCR2 promotes monocyte recruitment and intestinal inflammation in mice lacking the interleukin-10 receptor. Sci. Rep. 2022, 12, 452. [Google Scholar] [CrossRef]
  147. Platt, A.M.; Bain, C.C.; Bordon, Y.; Sester, D.P.; Mowat, A.M. An independent subset of TLR expressing CCR2-dependent macrophages promotes colonic inflammation. J. Immunol. 2010, 184, 6843–6854. [Google Scholar] [CrossRef] [Green Version]
  148. Andres, P.G.; Beck, P.L.; Mizoguchi, E.; Mizoguchi, A.; Bhan, A.K.; Dawson, T.; Kuziel, W.A.; Maeda, N.; MacDermott, R.P.; Podolsky, D.K.; et al. Mice with a selective deletion of the CC chemokine receptors 5 or 2 are protected from dextran sodium sulfate-mediated colitis: Lack of CC chemokine receptor 5 expression results in a NK1.1+ lymphocyte-associated Th2-type immune response in the intestine. J. Immunol. 2000, 164, 6303–6312. [Google Scholar] [CrossRef] [Green Version]
  149. Smythies, L.E.; Maheshwari, A.; Clements, R.; Eckhoff, D.; Novak, L.; Vu, H.L.; Mosteller-Barnum, L.M.; Sellers, M.; Smith, P.D. Mucosal IL-8 and TGF-beta recruit blood monocytes: Evidence for cross-talk between the lamina propria stroma and myeloid cells. J. Leukoc. Biol. 2006, 80, 492–499. [Google Scholar] [CrossRef] [Green Version]
  150. Redhu, N.S.; Bakthavatchalu, V.; Conaway, E.A.; Shouval, D.S.; Tsou, A.; Goettel, J.A.; Biswas, A.; Wang, C.; Field, M.; Muller, W.; et al. Macrophage dysfunction initiates colitis during weaning of infant mice lacking the interleukin-10 receptor. elife 2017, 6, e27652. [Google Scholar] [CrossRef]
  151. Rugtveit, J.; Nilsen, E.M.; Bakka, A.; Carlsen, H.; Brandtzaeg, P.; Scott, H. Cytokine profiles differ in newly recruited and resident subsets of mucosal macrophages from inflammatory bowel disease. Gastroenterology 1997, 112, 1493–1505. [Google Scholar] [CrossRef]
  152. Thiesen, S.; Janciauskiene, S.; Uronen-Hansson, H.; Agace, W.; Hogerkorp, C.M.; Spee, P.; Hakansson, K.; Grip, O. CD14(hi)HLA-DR(dim) macrophages, with a resemblance to classical blood monocytes, dominate inflamed mucosa in Crohn’s disease. J. Leukoc. Biol. 2014, 95, 531–541. [Google Scholar] [CrossRef] [PubMed]
  153. Perminow, G.; Reikvam, D.H.; Lyckander, L.G.; Brandtzaeg, P.; Vatn, M.H.; Carlsen, H.S. Increased number and activation of colonic macrophages in pediatric patients with untreated Crohn’s disease. Inflamm. Bowel Dis. 2009, 15, 1368–1378. [Google Scholar] [CrossRef]
  154. Grimm, M.C.; Pullman, W.E.; Bennett, G.M.; Sullivan, P.J.; Pavli, P.; Doe, W.F. Direct evidence of monocyte recruitment to inflammatory bowel disease mucosa. J. Gastroenterol. Hepatol. 1995, 10, 387–395. [Google Scholar] [CrossRef]
  155. Mantovani, A.; Biswas, S.K.; Galdiero, M.R.; Sica, A.; Locati, M. Macrophage plasticity and polarization in tissue repair and remodelling. J. Pathol. 2013, 229, 176–185. [Google Scholar] [CrossRef]
  156. Kostadinova, F.I.; Baba, T.; Ishida, Y.; Kondo, T.; Popivanova, B.K.; Mukaida, N. Crucial involvement of the CX3CR1-CX3CL1 axis in dextran sulfate sodium-mediated acute colitis in mice. J. Leukoc. Biol. 2010, 88, 133–143. [Google Scholar] [CrossRef]
  157. Brand, S.; Hofbauer, K.; Dambacher, J.; Schnitzler, F.; Staudinger, T.; Pfennig, S.; Seiderer, J.; Tillack, C.; Konrad, A.; Goke, B.; et al. Increased expression of the chemokine fractalkine in Crohn’s disease and association of the fractalkine receptor T280M polymorphism with a fibrostenosing disease Phenotype. Am. J. Gastroenterol. 2006, 101, 99–106. [Google Scholar] [CrossRef]
  158. Sabate, J.M.; Ameziane, N.; Lamoril, J.; Jouet, P.; Farmachidi, J.P.; Soule, J.C.; Harnois, F.; Sobhani, I.; Jian, R.; Deybach, J.C.; et al. The V249I polymorphism of the CX3CR1 gene is associated with fibrostenotic disease behavior in patients with Crohn’s disease. Eur. J. Gastroenterol. Hepatol. 2008, 20, 748–755. [Google Scholar] [CrossRef]
  159. Begue, B.; Verdier, J.; Rieux-Laucat, F.; Goulet, O.; Morali, A.; Canioni, D.; Hugot, J.P.; Daussy, C.; Verkarre, V.; Pigneur, B.; et al. Defective IL10 signaling defining a subgroup of patients with inflammatory bowel disease. Am. J. Gastroenterol. 2011, 106, 1544–1555. [Google Scholar] [CrossRef]
  160. Franke, A.; Balschun, T.; Karlsen, T.H.; Sventoraityte, J.; Nikolaus, S.; Mayr, G.; Domingues, F.S.; Albrecht, M.; Nothnagel, M.; Ellinghaus, D.; et al. Sequence variants in IL10, ARPC2 and multiple other loci contribute to ulcerative colitis susceptibility. Nat. Genet. 2008, 40, 1319–1323. [Google Scholar] [CrossRef]
  161. Glocker, E.O.; Kotlarz, D.; Boztug, K.; Gertz, E.M.; Schaffer, A.A.; Noyan, F.; Perro, M.; Diestelhorst, J.; Allroth, A.; Murugan, D.; et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N. Engl. J. Med. 2009, 361, 2033–2045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Kotlarz, D.; Beier, R.; Murugan, D.; Diestelhorst, J.; Jensen, O.; Boztug, K.; Pfeifer, D.; Kreipe, H.; Pfister, E.D.; Baumann, U.; et al. Loss of interleukin-10 signaling and infantile inflammatory bowel disease: Implications for diagnosis and therapy. Gastroenterology 2012, 143, 347–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Kuhn, R.; Lohler, J.; Rennick, D.; Rajewsky, K.; Muller, W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 1993, 75, 263–274. [Google Scholar] [CrossRef] [PubMed]
  164. Moran, C.J.; Walters, T.D.; Guo, C.H.; Kugathasan, S.; Klein, C.; Turner, D.; Wolters, V.M.; Bandsma, R.H.; Mouzaki, M.; Zachos, M.; et al. IL-10R polymorphisms are associated with very-early-onset ulcerative colitis. Inflamm. Bowel Dis. 2013, 19, 115–123. [Google Scholar] [CrossRef] [PubMed]
  165. Spencer, S.D.; Di Marco, F.; Hooley, J.; Pitts-Meek, S.; Bauer, M.; Ryan, A.M.; Sordat, B.; Gibbs, V.C.; Aguet, M. The orphan receptor CRF2-4 is an essential subunit of the interleukin 10 receptor. J. Exp. Med. 1998, 187, 571–578. [Google Scholar] [CrossRef] [Green Version]
  166. Shouval, D.S.; Biswas, A.; Goettel, J.A.; McCann, K.; Conaway, E.; Redhu, N.S.; Mascanfroni, I.D.; Al Adham, Z.; Lavoie, S.; Ibourk, M.; et al. Interleukin-10 receptor signaling in innate immune cells regulates mucosal immune tolerance and anti-inflammatory macrophage function. Immunity 2014, 40, 706–719. [Google Scholar] [CrossRef] [Green Version]
  167. Bernshtein, B.; Curato, C.; Ioannou, M.; Thaiss, C.A.; Gross-Vered, M.; Kolesnikov, M.; Wang, Q.; David, E.; Chappell-Maor, L.; Harmelin, A.; et al. IL-23-producing IL-10Ralpha-deficient gut macrophages elicit an IL-22-driven proinflammatory epithelial cell response. Sci. Immunol. 2019, 4, eaau6571. [Google Scholar] [CrossRef]
  168. Asano, K.; Takahashi, N.; Ushiki, M.; Monya, M.; Aihara, F.; Kuboki, E.; Moriyama, S.; Iida, M.; Kitamura, H.; Qiu, C.H.; et al. Intestinal CD169(+) macrophages initiate mucosal inflammation by secreting CCL8 that recruits inflammatory monocytes. Nat. Commun. 2015, 6, 7802. [Google Scholar] [CrossRef] [Green Version]
  169. Daley, J.M.; Brancato, S.K.; Thomay, A.A.; Reichner, J.S.; Albina, J.E. The phenotype of murine wound macrophages. J. Leukoc. Biol. 2010, 87, 59–67. [Google Scholar] [CrossRef] [Green Version]
  170. Pull, S.L.; Doherty, J.M.; Mills, J.C.; Gordon, J.I.; Stappenbeck, T.S. Activated macrophages are an adaptive element of the colonic epithelial progenitor niche necessary for regenerative responses to injury. Proc. Natl. Acad. Sci. USA 2005, 102, 99–104. [Google Scholar] [CrossRef]
  171. Morhardt, T.L.; Hayashi, A.; Ochi, T.; Quiros, M.; Kitamoto, S.; Nagao-Kitamoto, H.; Kuffa, P.; Atarashi, K.; Honda, K.; Kao, J.Y.; et al. IL-10 produced by macrophages regulates epithelial integrity in the small intestine. Sci. Rep. 2019, 9, 1223. [Google Scholar] [CrossRef] [Green Version]
  172. Quiros, M.; Nishio, H.; Neumann, P.A.; Siuda, D.; Brazil, J.C.; Azcutia, V.; Hilgarth, R.; O’Leary, M.N.; Garcia-Hernandez, V.; Leoni, G.; et al. Macrophage-derived IL-10 mediates mucosal repair by epithelial WISP-1 signaling. J. Clin. Investig. 2017, 127, 3510–3520. [Google Scholar] [CrossRef] [Green Version]
  173. D’Angelo, F.; Bernasconi, E.; Schafer, M.; Moyat, M.; Michetti, P.; Maillard, M.H.; Velin, D. Macrophages promote epithelial repair through hepatocyte growth factor secretion. Clin. Exp. Immunol. 2013, 174, 60–72. [Google Scholar] [CrossRef]
  174. Ma, S.; Zhang, J.; Liu, H.; Li, S.; Wang, Q. The Role of Tissue-Resident Macrophages in the Development and Treatment of Inflammatory Bowel Disease. Front. Cell Dev. Biol. 2022, 10, 896591. [Google Scholar] [CrossRef]
  175. Cosin-Roger, J.; Ortiz-Masia, D.; Calatayud, S.; Hernandez, C.; Esplugues, J.V.; Barrachina, M.D. The activation of Wnt signaling by a STAT6-dependent macrophage phenotype promotes mucosal repair in murine IBD. Mucosal Immunol. 2016, 9, 986–998. [Google Scholar] [CrossRef] [Green Version]
  176. Seo, D.H.; Che, X.; Kwak, M.S.; Kim, S.; Kim, J.H.; Ma, H.W.; Kim, D.H.; Kim, T.I.; Kim, W.H.; Kim, S.W.; et al. Interleukin-33 regulates intestinal inflammation by modulating macrophages in inflammatory bowel disease. Sci. Rep. 2017, 7, 851. [Google Scholar] [CrossRef] [Green Version]
  177. Waddell, A.; Vallance, J.E.; Moore, P.D.; Hummel, A.T.; Wu, D.; Shanmukhappa, S.K.; Fei, L.; Washington, M.K.; Minar, P.; Coburn, L.A.; et al. IL-33 Signaling Protects from Murine Oxazolone Colitis by Supporting Intestinal Epithelial Function. Inflamm. Bowel Dis. 2015, 21, 2737–2746. [Google Scholar] [CrossRef] [Green Version]
  178. Schleier, L.; Wiendl, M.; Heidbreder, K.; Binder, M.T.; Atreya, R.; Rath, T.; Becker, E.; Schulz-Kuhnt, A.; Stahl, A.; Schulze, L.L.; et al. Non-classical monocyte homing to the gut via alpha4beta7 integrin mediates macrophage-dependent intestinal wound healing. Gut 2020, 69, 252–263. [Google Scholar] [CrossRef]
  179. Pakshir, P.; Hinz, B. The big five in fibrosis: Macrophages, myofibroblasts, matrix, mechanics, and miscommunication. Matrix Biol. 2018, 68–69, 81–93. [Google Scholar] [CrossRef]
  180. Wynn, T.A.; Barron, L. Macrophages: Master regulators of inflammation and fibrosis. Semin. Liver Dis. 2010, 30, 245–257. [Google Scholar] [CrossRef]
  181. Wynn, T.A.; Ramalingam, T.R. Mechanisms of fibrosis: Therapeutic translation for fibrotic disease. Nat. Med. 2012, 18, 1028–1040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Lenti, M.V.; Di Sabatino, A. Intestinal fibrosis. Mol. Aspects Med. 2019, 65, 100–109. [Google Scholar] [CrossRef] [PubMed]
  183. D’Alessio, S.; Ungaro, F.; Noviello, D.; Lovisa, S.; Peyrin-Biroulet, L.; Danese, S. Revisiting fibrosis in inflammatory bowel disease: The gut thickens. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 169–184. [Google Scholar] [CrossRef] [PubMed]
  184. Hinz, B.; Lagares, D. Evasion of apoptosis by myofibroblasts: A hallmark of fibrotic diseases. Nat. Rev. Rheumatol. 2020, 16, 11–31. [Google Scholar] [CrossRef]
  185. Franze, E.; Dinallo, V.; Laudisi, F.; Di Grazia, A.; Di Fusco, D.; Colantoni, A.; Ortenzi, A.; Giuffrida, P.; Di Carlo, S.; Sica, G.S.; et al. Interleukin-34 Stimulates Gut Fibroblasts to Produce Collagen Synthesis. J. Crohn’s Colitis 2020, 14, 1436–1445. [Google Scholar] [CrossRef]
  186. Cupedo, T.; Crellin, N.K.; Papazian, N.; Rombouts, E.J.; Weijer, K.; Grogan, J.L.; Fibbe, W.E.; Cornelissen, J.J.; Spits, H. Human fetal lymphoid tissue-inducer cells are interleukin 17-producing precursors to RORC+ CD127+ natural killer-like cells. Nat. Immunol. 2009, 10, 66–74. [Google Scholar] [CrossRef]
  187. Cella, M.; Fuchs, A.; Vermi, W.; Facchetti, F.; Otero, K.; Lennerz, J.K.; Doherty, J.M.; Mills, J.C.; Colonna, M. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature 2009, 457, 722–725. [Google Scholar] [CrossRef] [Green Version]
  188. Spits, H.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.; Mebius, R.E.; et al. Innate lymphoid cells—A proposal for uniform nomenclature. Nat. Rev. Immunol. 2013, 13, 145–149. [Google Scholar] [CrossRef]
  189. Ignacio, A.; Breda, C.N.S.; Camara, N.O.S. Innate lymphoid cells in tissue homeostasis and diseases. World. J. Hepatol. 2017, 9, 979–989. [Google Scholar] [CrossRef]
  190. Eberl, G.; Colonna, M.; Di Santo, J.P.; McKenzie, A.N. Innate lymphoid cells. Innate lymphoid cells: A new paradigm in immunology. Science 2015, 348, aaa6566. [Google Scholar] [CrossRef]
  191. Artis, D.; Spits, H. The biology of innate lymphoid cells. Nature 2015, 517, 293–301. [Google Scholar] [CrossRef]
  192. Huang, Y.; Mao, K.; Germain, R.N. Thinking differently about ILCs-Not just tissue resident and not just the same as CD4(+) T-cell effectors. Immunol. Rev. 2018, 286, 160–171. [Google Scholar] [CrossRef]
  193. Trabanelli, S.; Gomez-Cadena, A.; Salome, B.; Michaud, K.; Mavilio, D.; Landis, B.N.; Jandus, P.; Jandus, C. Human innate lymphoid cells (ILCs): Toward a uniform immune-phenotyping. Cytom. B Clin. Cytom. 2018, 94, 392–399. [Google Scholar] [CrossRef]
  194. Panda, S.K.; Colonna, M. Innate Lymphoid Cells in Mucosal Immunity. Front. Immunol. 2019, 10, 861. [Google Scholar] [CrossRef] [Green Version]
  195. Giuffrida, P.; Corazza, G.R.; Di Sabatino, A. Old and New Lymphocyte Players in Inflammatory Bowel Disease. Dig. Dis. Sci. 2018, 63, 277–288. [Google Scholar] [CrossRef]
  196. Sepahi, A.; Liu, Q.; Friesen, L.; Kim, C.H. Dietary fiber metabolites regulate innate lymphoid cell responses. Mucosal Immunol. 2021, 14, 317–330. [Google Scholar] [CrossRef]
  197. Luo, W.; Tian, L.; Tan, B.; Shen, Z.; Xiao, M.; Wu, S.; Meng, X.; Wu, X.; Wang, X. Update: Innate Lymphoid Cells in Inflammatory Bowel Disease. Dig. Dis. Sci. 2021, 67, 56–66. [Google Scholar] [CrossRef]
  198. Zhou, W.; Sonnenberg, G.F. Activation and Suppression of Group 3 Innate Lymphoid Cells in the Gut. Trends Immunol. 2020, 41, 721–733. [Google Scholar] [CrossRef]
  199. Wu, Y.; Shen, J. Innate Lymphoid Cells in Crohn’s Disease. Front. Immunol. 2020, 11, 554880. [Google Scholar] [CrossRef]
  200. Diefenbach, A.; Gnafakis, S.; Shomrat, O. Innate Lymphoid Cell-Epithelial Cell Modules Sustain Intestinal Homeostasis. Immunity 2020, 52, 452–463. [Google Scholar] [CrossRef]
  201. Saez, A.; Gomez-Bris, R.; Herrero-Fernandez, B.; Mingorance, C.; Rius, C.; Gonzalez-Granado, J.M. Innate Lymphoid Cells in Intestinal Homeostasis and Inflammatory Bowel Disease. Int. J. Mol. Sci. 2021, 22, 7618. [Google Scholar] [CrossRef] [PubMed]
  202. Zeng, B.; Shi, S.; Ashworth, G.; Dong, C.; Liu, J.; Xing, F. ILC3 function as a double-edged sword in inflammatory bowel diseases. Cell Death Dis. 2019, 10, 315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Schulz-Kuhnt, A.; Neurath, M.F.; Wirtz, S.; Atreya, I. Innate Lymphoid Cells as Regulators of Epithelial Integrity: Therapeutic Implications for Inflammatory Bowel Diseases. Front. Med. 2021, 8, 656745. [Google Scholar] [CrossRef] [PubMed]
  204. Peng, V.; Jaeger, N.; Colonna, M. Innate Lymphoid Cells and Inflammatory Bowel Disease. Adv. Exp. Med. Biol. 2022, 1365, 97–112. [Google Scholar] [PubMed]
  205. Schraml, B.U.; Reis e Sousa, C. Defining dendritic cells. Curr. Opin. Immunol. 2015, 32, 13–20. [Google Scholar] [CrossRef] [Green Version]
  206. Mildner, A.; Jung, S. Development and function of dendritic cell subsets. Immunity 2014, 40, 642–656. [Google Scholar] [CrossRef] [Green Version]
  207. Guilliams, M.; Dutertre, C.A.; Scott, C.L.; McGovern, N.; Sichien, D.; Chakarov, S.; Van Gassen, S.; Chen, J.; Poidinger, M.; De Prijck, S.; et al. Unsupervised High-Dimensional Analysis Aligns Dendritic Cells across Tissues and Species. Immunity 2016, 45, 669–684. [Google Scholar] [CrossRef] [Green Version]
  208. Dutertre, C.A.; Wang, L.F.; Ginhoux, F. Aligning bona fide dendritic cell populations across species. Cell. Immunol. 2014, 291, 3–10. [Google Scholar] [CrossRef]
  209. Zernecke, A. Dendritic cells in atherosclerosis: Evidence in mice and humans. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 763–770. [Google Scholar] [CrossRef] [Green Version]
  210. Gil-Pulido, J.; Zernecke, A. Antigen-presenting dendritic cells in atherosclerosis. Eur. J. Pharmacol. 2017, 816, 25–31. [Google Scholar] [CrossRef]
  211. Greter, M.; Helft, J.; Chow, A.; Hashimoto, D.; Mortha, A.; Agudo-Cantero, J.; Bogunovic, M.; Gautier, E.L.; Miller, J.; Leboeuf, M.; et al. GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells. Immunity 2012, 36, 1031–1046. [Google Scholar] [CrossRef] [Green Version]
  212. Ushach, I.; Zlotnik, A. Biological role of granulocyte macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) on cells of the myeloid lineage. J. Leukoc. Biol. 2016, 100, 481–489. [Google Scholar] [CrossRef] [Green Version]
  213. Banchereau, J.; Briere, F.; Caux, C.; Davoust, J.; Lebecque, S.; Liu, Y.J.; Pulendran, B.; Palucka, K. Immunobiology of dendritic cells. Annu. Rev. Immunol. 2000, 18, 767–811. [Google Scholar] [CrossRef]
  214. Mellman, I.; Steinman, R.M. Dendritic cells: Specialized and regulated antigen processing machines. Cell 2001, 106, 255–258. [Google Scholar] [CrossRef] [Green Version]
  215. Palucka, K.; Banchereau, J. Dendritic cells: A link between innate and adaptive immunity. J. Clin. Immunol. 1999, 19, 12–25. [Google Scholar] [CrossRef]
  216. Palucka, K.; Banchereau, J. Linking innate and adaptive immunity. Nat. Med. 1999, 5, 868–870. [Google Scholar] [CrossRef]
  217. Bousso, P. T-cell activation by dendritic cells in the lymph node: Lessons from the movies. Nat. Rev. Immunol. 2008, 8, 675–684. [Google Scholar] [CrossRef]
  218. Steinman, R.M. Decisions about dendritic cells: Past, present, and future. Annu. Rev. Immunol. 2012, 30, 1–22. [Google Scholar] [CrossRef] [Green Version]
  219. Lelouard, H.; Fallet, M.; de Bovis, B.; Meresse, S.; Gorvel, J.P. Peyer’s patch dendritic cells sample antigens by extending dendrites through M cell-specific transcellular pores. Gastroenterology 2012, 142, 592–601.e3. [Google Scholar] [CrossRef]
  220. Ohta, T.; Sugiyama, M.; Hemmi, H.; Yamazaki, C.; Okura, S.; Sasaki, I.; Fukuda, Y.; Orimo, T.; Ishii, K.J.; Hoshino, K.; et al. Crucial roles of XCR1-expressing dendritic cells and the XCR1-XCL1 chemokine axis in intestinal immune homeostasis. Sci. Rep. 2016, 6, 23505. [Google Scholar] [CrossRef]
  221. Johansson-Lindbom, B.; Svensson, M.; Pabst, O.; Palmqvist, C.; Marquez, G.; Forster, R.; Agace, W.W. Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing. J. Exp. Med. 2005, 202, 1063–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Agace, W.W.; Persson, E.K. How vitamin A metabolizing dendritic cells are generated in the gut mucosa. Trends Immunol. 2012, 33, 42–48. [Google Scholar] [CrossRef] [PubMed]
  223. Ohtani, M.; Hoshii, T.; Fujii, H.; Koyasu, S.; Hirao, A.; Matsuda, S. Cutting edge: mTORC1 in intestinal CD11c+ CD11b+ dendritic cells regulates intestinal homeostasis by promoting IL-10 production. J. Immunol. 2012, 188, 4736–4740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Al-Hassi, H.O.; Mann, E.R.; Sanchez, B.; English, N.R.; Peake, S.T.; Landy, J.; Man, R.; Urdaci, M.; Hart, A.L.; Fernandez-Salazar, L.; et al. Altered human gut dendritic cell properties in ulcerative colitis are reversed by Lactobacillus plantarum extracellular encrypted peptide STp. Mol. Nutr. Food Res. 2014, 58, 1132–1143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Hart, A.L.; Al-Hassi, H.O.; Rigby, R.J.; Bell, S.J.; Emmanuel, A.V.; Knight, S.C.; Kamm, M.A.; Stagg, A.J. Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology 2005, 129, 50–65. [Google Scholar] [CrossRef] [PubMed]
  226. Magnusson, M.K.; Brynjolfsson, S.F.; Dige, A.; Uronen-Hansson, H.; Borjesson, L.G.; Bengtsson, J.L.; Gudjonsson, S.; Ohman, L.; Agnholt, J.; Sjovall, H.; et al. Macrophage and dendritic cell subsets in IBD: ALDH+ cells are reduced in colon tissue of patients with ulcerative colitis regardless of inflammation. Mucosal Immunol. 2016, 9, 171–182. [Google Scholar] [CrossRef] [Green Version]
  227. Faleiro, R.; Liu, J.; Karunarathne, D.; Edmundson, A.; Winterford, C.; Nguyen, T.H.; Simms, L.A.; Radford-Smith, G.; Wykes, M. Crohn’s disease is facilitated by a disturbance of programmed death-1 ligand 2 on blood dendritic cells. Clin. Transl. Immunology 2019, 8, e01071. [Google Scholar] [CrossRef]
  228. Sawai, C.M.; Serpas, L.; Neto, A.G.; Jang, G.; Rashidfarrokhi, A.; Kolbeck, R.; Sanjuan, M.A.; Reizis, B.; Sisirak, V. Plasmacytoid Dendritic Cells Are Largely Dispensable for the Pathogenesis of Experimental Inflammatory Bowel Disease. Front. Immunol. 2018, 9, 2475. [Google Scholar] [CrossRef]
  229. Mishima, Y.; Sartor, R.B. Manipulating resident microbiota to enhance regulatory immune function to treat inflammatory bowel diseases. J. Gastroenterol. 2020, 55, 4–14. [Google Scholar] [CrossRef] [Green Version]
  230. Xavier, R.J.; Podolsky, D.K. Unravelling the pathogenesis of inflammatory bowel disease. Nature 2007, 448, 427–434. [Google Scholar] [CrossRef]
  231. Matsuno, H.; Kayama, H.; Nishimura, J.; Sekido, Y.; Osawa, H.; Barman, S.; Ogino, T.; Takahashi, H.; Haraguchi, N.; Hata, T.; et al. CD103+ Dendritic Cell Function Is Altered in the Colons of Patients with Ulcerative Colitis. Inflamm. Bowel Dis. 2017, 23, 1524–1534. [Google Scholar] [CrossRef] [Green Version]
  232. Pool, L.; Rivollier, A.; Agace, W.W. Deletion of IRF4 in Dendritic Cells Leads to Delayed Onset of T Cell-Dependent Colitis. J. Immunol. 2020, 204, 1047–1055. [Google Scholar] [CrossRef]
  233. Muzaki, A.R.; Tetlak, P.; Sheng, J.; Loh, S.C.; Setiagani, Y.A.; Poidinger, M.; Zolezzi, F.; Karjalainen, K.; Ruedl, C. Intestinal CD103(+)CD11b(-) dendritic cells restrain colitis via IFN-gamma-induced anti-inflammatory response in epithelial cells. Mucosal Immunol. 2016, 9, 336–351. [Google Scholar] [CrossRef] [Green Version]
  234. Welty, N.E.; Staley, C.; Ghilardi, N.; Sadowsky, M.J.; Igyarto, B.Z.; Kaplan, D.H. Intestinal lamina propria dendritic cells maintain T cell homeostasis but do not affect commensalism. J. Exp. Med. 2013, 210, 2011–2024. [Google Scholar] [CrossRef] [Green Version]
  235. Furey, T.S.; Sethupathy, P.; Sheikh, S.Z. Redefining the IBDs using genome-scale molecular phenotyping. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 296–311. [Google Scholar] [CrossRef]
  236. Graham, D.B.; Xavier, R.J. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature 2020, 578, 527–539. [Google Scholar] [CrossRef]
  237. De Lange, K.M.; Moutsianas, L.; Lee, J.C.; Lamb, C.A.; Luo, Y.; Kennedy, N.A.; Jostins, L.; Rice, D.L.; Gutierrez-Achury, J.; Ji, S.G.; et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat. Genet. 2017, 49, 256–261. [Google Scholar] [CrossRef] [Green Version]
  238. Loddo, I.; Romano, C. Inflammatory Bowel Disease: Genetics, Epigenetics, and Pathogenesis. Front. Immunol. 2015, 6, 551. [Google Scholar] [CrossRef] [Green Version]
  239. Jostins, L.; Ripke, S.; Weersma, R.K.; Duerr, R.H.; McGovern, D.P.; Hui, K.Y.; Lee, J.C.; Schumm, L.P.; Sharma, Y.; Anderson, C.A.; et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012, 491, 119–124. [Google Scholar] [CrossRef] [Green Version]
  240. Mitsialis, V.; Wall, S.; Liu, P.; Ordovas-Montanes, J.; Parmet, T.; Vukovic, M.; Spencer, D.; Field, M.; McCourt, C.; Toothaker, J.; et al. Single-Cell Analyses of Colon and Blood Reveal Distinct Immune Cell Signatures of Ulcerative Colitis and Crohn’s Disease. Gastroenterology 2020, 159, 591–608.e10. [Google Scholar] [CrossRef]
  241. Martin, J.C.; Chang, C.; Boschetti, G.; Ungaro, R.; Giri, M.; Grout, J.A.; Gettler, K.; Chuang, L.S.; Nayar, S.; Greenstein, A.J.; et al. Single-Cell Analysis of Crohn’s Disease Lesions Identifies a Pathogenic Cellular Module Associated with Resistance to Anti-TNF Therapy. Cell 2019, 178, 1493–1508.e20. [Google Scholar] [CrossRef] [PubMed]
  242. Corridoni, D.; Antanaviciute, A.; Gupta, T.; Fawkner-Corbett, D.; Aulicino, A.; Jagielowicz, M.; Parikh, K.; Repapi, E.; Taylor, S.; Ishikawa, D.; et al. Single-cell atlas of colonic CD8(+) T cells in ulcerative colitis. Nat. Med. 2020, 26, 1480–1490. [Google Scholar] [CrossRef] [PubMed]
  243. Boland, B.S.; He, Z.; Tsai, M.S.; Olvera, J.G.; Omilusik, K.D.; Duong, H.G.; Kim, E.S.; Limary, A.E.; Jin, W.; Milner, J.J.; et al. Heterogeneity and clonal relationships of adaptive immune cells in ulcerative colitis revealed by single-cell analyses. Sci. Immunol. 2020, 5, eabb4432. [Google Scholar] [CrossRef] [PubMed]
  244. Smillie, C.S.; Biton, M.; Ordovas-Montanes, J.; Sullivan, K.M.; Burgin, G.; Graham, D.B.; Herbst, R.H.; Rogel, N.; Slyper, M.; Waldman, J.; et al. Intra- and Inter-cellular Rewiring of the Human Colon during Ulcerative Colitis. Cell 2019, 178, 714–730.e22. [Google Scholar] [CrossRef] [PubMed]
  245. Zhang, M.J.; Hou, K.; Dey, K.K.; Sakaue, S.; Jagadeesh, K.A.; Weinand, K.; Taychameekiatchai, A.; Rao, P.; Pisco, A.O.; Zou, J.; et al. Polygenic enrichment distinguishes disease associations of individual cells in single-cell RNA-seq data. Nat. Genet. 2022, 54, 1572–1580. [Google Scholar] [CrossRef]
  246. Saul, D.; Leite Barros, L.; Wixom, A.Q.; Gellhaus, B.; Gibbons, H.R.; Faubion, W.A.; Kosinsky, R.L. Cell Type-Specific Induction of Inflammation-Associated Genes in Crohn’s Disease and Colorectal Cancer. Int. J. Mol. Sci. 2022, 23, 3082. [Google Scholar] [CrossRef]
  247. Li, G.; Zhang, B.; Hao, J.; Chu, X.; Wiestler, M.; Cornberg, M.; Xu, C.J.; Liu, X.; Li, Y. Identification of Novel Population-Specific Cell Subsets in Chinese Ulcerative Colitis Patients Using Single-Cell RNA Sequencing. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 99–117. [Google Scholar] [CrossRef]
  248. Zhao, Q.; Zhang, T.; Yang, H. ScRNA-seq identified the metabolic reprogramming of human colonic immune cells in different locations and disease states. Biochem. Biophys. Res. Commun. 2022, 604, 96–103. [Google Scholar] [CrossRef]
  249. Diez-Obrero, V.; Moratalla-Navarro, F.; Ibanez-Sanz, G.; Guardiola, J.; Rodriguez-Moranta, F.; Obon-Santacana, M.; Diez-Villanueva, A.; Dampier, C.H.; Devall, M.; Carreras-Torres, R.; et al. Transcriptome-Wide Association Study for Inflammatory Bowel Disease Reveals Novel Candidate Susceptibility Genes in Specific Colon Subsites and Tissue Categories. J. Crohn’s Colitis 2022, 16, 275–285. [Google Scholar] [CrossRef]
  250. Valenzuela, P.L.; Amo, C.; Sanchez-Martinez, G.; Torrontegi, E.; Vazquez-Carrion, J.; Montalvo, Z.; Lucia, A.; de la Villa, P. Enhancement of Mood but not Performance in Elite Athletes With Transcranial Direct-Current Stimulation. Int. J. Sports Physiol. Perform. 2019, 14, 310–316. [Google Scholar] [CrossRef]
  251. Fukunaga, S.; Kuwaki, K.; Mitsuyama, K.; Takedatsu, H.; Yoshioka, S.; Yamasaki, H.; Yamauchi, R.; Mori, A.; Kakuma, T.; Tsuruta, O.; et al. Detection of calprotectin in inflammatory bowel disease: Fecal and serum levels and immunohistochemical localization. Int. J. Mol. Med. 2018, 41, 107–118. [Google Scholar] [CrossRef] [Green Version]
  252. Muthas, D.; Reznichenko, A.; Balendran, C.A.; Bottcher, G.; Clausen, I.G.; Karrman Mardh, C.; Ottosson, T.; Uddin, M.; MacDonald, T.T.; Danese, S.; et al. Neutrophils in ulcerative colitis: A review of selected biomarkers and their potential therapeutic implications. Scand. J. Gastroenterol. 2017, 52, 125–135. [Google Scholar] [CrossRef]
  253. Barry, R.; Ruano-Gallego, D.; Radhakrishnan, S.T.; Lovell, S.; Yu, L.; Kotik, O.; Glegola-Madejska, I.; Tate, E.W.; Choudhary, J.S.; Williams, H.R.T.; et al. Faecal neutrophil elastase-antiprotease balance reflects colitis severity. Mucosal Immunol. 2020, 13, 322–333. [Google Scholar] [CrossRef] [Green Version]
  254. Jablaoui, A.; Kriaa, A.; Mkaouar, H.; Akermi, N.; Soussou, S.; Wysocka, M.; Woloszyn, D.; Amouri, A.; Gargouri, A.; Maguin, E.; et al. Fecal Serine Protease Profiling in Inflammatory Bowel Diseases. Front. Cell. Infect. Microbiol. 2020, 10, 21. [Google Scholar] [CrossRef]
  255. Kumar, K.G.; Trevaskis, J.L.; Lam, D.D.; Sutton, G.M.; Koza, R.A.; Chouljenko, V.N.; Kousoulas, K.G.; Rogers, P.M.; Kesterson, R.A.; Thearle, M.; et al. Identification of adropin as a secreted factor linking dietary macronutrient intake with energy homeostasis and lipid metabolism. Cell Metab. 2008, 8, 468–481. [Google Scholar] [CrossRef] [Green Version]
  256. Simac, P.; Perkovic, D.; Bozic, I.; Bilopavlovic, N.; Martinovic, D.; Bozic, J. Serum Adropin Levels in Patients with Rheumatoid Arthritis. Life 2022, 12, 169. [Google Scholar] [CrossRef]
  257. Brnic, D.; Martinovic, D.; Zivkovic, P.M.; Tokic, D.; Tadin Hadjina, I.; Rusic, D.; Vilovic, M.; Supe-Domic, D.; Tonkic, A.; Bozic, J. Serum adropin levels are reduced in patients with inflammatory bowel diseases. Sci. Rep. 2020, 10, 9264. [Google Scholar] [CrossRef]
  258. Zivkovic, P.M.; Matetic, A.; Tadin Hadjina, I.; Rusic, D.; Vilovic, M.; Supe-Domic, D.; Borovac, J.A.; Mudnic, I.; Tonkic, A.; Bozic, J. Serum Catestatin Levels and Arterial Stiffness Parameters Are Increased in Patients with Inflammatory Bowel Disease. J. Clin. Med. 2020, 9, 628. [Google Scholar] [CrossRef] [Green Version]
  259. Muntjewerff, E.M.; Tang, K.; Lutter, L.; Christoffersson, G.; Nicolasen, M.J.T.; Gao, H.; Katkar, G.D.; Das, S.; Ter Beest, M.; Ying, W.; et al. Chromogranin A regulates gut permeability via the antagonistic actions of its proteolytic peptides. Acta Physiol. 2021, 232, e13655. [Google Scholar] [CrossRef]
  260. Rabbi, M.F.; Labis, B.; Metz-Boutigue, M.H.; Bernstein, C.N.; Ghia, J.E. Catestatin decreases macrophage function in two mouse models of experimental colitis. Biochem. Pharmacol. 2014, 89, 386–398. [Google Scholar] [CrossRef]
  261. Mahata, S.K.; Mahata, M.; Fung, M.M.; O’Connor, D.T. Catestatin: A multifunctional peptide from chromogranin A. Regul. Pept. 2010, 162, 33–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Khan, W.I.; Ghia, J.E. Gut hormones: Emerging role in immune activation and inflammation. Clin. Exp. Immunol. 2010, 161, 19–27. [Google Scholar] [CrossRef] [PubMed]
  263. Eissa, N.; Hussein, H.; Mesgna, R.; Bonin, S.; Hendy, G.N.; Metz-Boutigue, M.H.; Bernstein, C.N.; Ghia, J.E. Catestatin Regulates Epithelial Cell Dynamics to Improve Intestinal Inflammation. Vaccines 2018, 6, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Rabbi, M.F.; Munyaka, P.M.; Eissa, N.; Metz-Boutigue, M.H.; Khafipour, E.; Ghia, J.E. Human Catestatin Alters Gut Microbiota Composition in Mice. Front. Microbiol. 2016, 7, 2151. [Google Scholar] [CrossRef] [Green Version]
  265. Zigmond, E.; Jung, S. Intestinal macrophages: Well educated exceptions from the rule. Trends Immunol. 2013, 34, 162–168. [Google Scholar] [CrossRef]
  266. Yona, S.; Kim, K.W.; Wolf, Y.; Mildner, A.; Varol, D.; Breker, M.; Strauss-Ayali, D.; Viukov, S.; Guilliams, M.; Misharin, A.; et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 2013, 38, 79–91. [Google Scholar] [CrossRef] [Green Version]
  267. Gisbert, J.P.; Chaparro, M. Primary Failure to an Anti-TNF Agent in Inflammatory Bowel Disease: Switch (to a Second Anti-TNF Agent) or Swap (for Another Mechanism of Action)? J. Clin. Med. 2021, 10, 5318. [Google Scholar] [CrossRef]
  268. Yanai, H.; Hanauer, S.B. Assessing response and loss of response to biological therapies in IBD. Am. J. Gastroenterol. 2011, 106, 685–698. [Google Scholar] [CrossRef]
Figure 1. In healthy intestine (left), damage to the intestinal barrier triggers the recruitment of neutrophils from the circulation to the inflamed tissue along a chemotactic gradient formed by cytokines (IL-1β, IL-6, TNF-α), chemokines (CCL8, CXCL10, MIP-2), and growth factors (GM-CSF, G-CSF). Neutrophil recruitment is also mediated by bacteria-derived molecules such as formyl-methionyl-leucyl-phenylalanine (fMLP) and short-chain fatty acids (SCFAs). The recruited neutrophils participate in the elimination of microorganisms through phagocytosis, degranulation, reactive oxygen species (ROS) generation, and the release of neutrophil extracellular traps (NETs). Once their functions are completed, neutrophils undergo apoptosis and efferocytosis, facilitating the resolution of inflammation, tissue repair, and a return to normal tissue homeostasis. The participation of neutrophils and NETs in IBD is a double-edged sword (right). Neutrophils cooperate in wound healing and the resolution of inflammation by releasing vascular endothelial growth factors (VEGFs) and lipid mediators (protectin D1, resolvin E1). These factors impede neutrophil recruitment and promote phagocytosis. NETs impede the spread of microorganisms by trapping them in an environment of microbicidal components and stimulate the healing of the intestinal mucosa. Neutrophils directly cause tissue damage by releasing neutrophil elastase, proteases (MMPs), pro-inflammatory cytokines (IL-8, TNF-α, IL-1β), leukotriene B4, and ROS. These factors provoke not only injury to the epithelial barrier, but also the recruitment of neutrophils and other immune cells to the inflamed tissue. Neutrophil recruitment is also promoted by the cytokines IL-1α, IL-17, IL-22, G-CSF, and GM-CSF. Lack of the IBD protective gene CARD9 in neutrophils enhances ROS generation. IL-8, TNF-α, and PAD4 (increased in UC patients) contribute to NET production. Accumulation of NETs in the colon is accompanied by the induction of tissue damage and inflammation, as NETs also boost TNF-α and IL-1β production. Part of the figure was generated by using pictures from Servier Medical Art.
Figure 1. In healthy intestine (left), damage to the intestinal barrier triggers the recruitment of neutrophils from the circulation to the inflamed tissue along a chemotactic gradient formed by cytokines (IL-1β, IL-6, TNF-α), chemokines (CCL8, CXCL10, MIP-2), and growth factors (GM-CSF, G-CSF). Neutrophil recruitment is also mediated by bacteria-derived molecules such as formyl-methionyl-leucyl-phenylalanine (fMLP) and short-chain fatty acids (SCFAs). The recruited neutrophils participate in the elimination of microorganisms through phagocytosis, degranulation, reactive oxygen species (ROS) generation, and the release of neutrophil extracellular traps (NETs). Once their functions are completed, neutrophils undergo apoptosis and efferocytosis, facilitating the resolution of inflammation, tissue repair, and a return to normal tissue homeostasis. The participation of neutrophils and NETs in IBD is a double-edged sword (right). Neutrophils cooperate in wound healing and the resolution of inflammation by releasing vascular endothelial growth factors (VEGFs) and lipid mediators (protectin D1, resolvin E1). These factors impede neutrophil recruitment and promote phagocytosis. NETs impede the spread of microorganisms by trapping them in an environment of microbicidal components and stimulate the healing of the intestinal mucosa. Neutrophils directly cause tissue damage by releasing neutrophil elastase, proteases (MMPs), pro-inflammatory cytokines (IL-8, TNF-α, IL-1β), leukotriene B4, and ROS. These factors provoke not only injury to the epithelial barrier, but also the recruitment of neutrophils and other immune cells to the inflamed tissue. Neutrophil recruitment is also promoted by the cytokines IL-1α, IL-17, IL-22, G-CSF, and GM-CSF. Lack of the IBD protective gene CARD9 in neutrophils enhances ROS generation. IL-8, TNF-α, and PAD4 (increased in UC patients) contribute to NET production. Accumulation of NETs in the colon is accompanied by the induction of tissue damage and inflammation, as NETs also boost TNF-α and IL-1β production. Part of the figure was generated by using pictures from Servier Medical Art.
Ijms 24 01526 g001
Figure 2. Macrophages and dendritic cell in homeostasis and IBD. Macrophages and DCs play important roles in homeostasis and in the development of IBD by phagocytosing cellular debris, producing cytokines, regulating tissue repair, and interacting with other cells. In the intestine, monocyte-derived macrophages (MΦ) are more abundant than tissue-resident macrophages of embryonic origin. Both perform phagocytosis, produce cytokines, and interact with other cells. Upon weaning, bone marrow-derived monocytes egress from the circulation and extravasate into the tissue, where they undergo differentiation and maturation (downregulation of Ly6C, production of MHCII, and increased expression of CX3CR1). In homeostasis, tissue-resident macrophages in the muscularis externa interact with enteric and myenteric neurons controlling intestinal secretion and motility, while in the lamina propria, macrophages provide signals to intestinal stem cells that give rise to goblet cells, Paneth cells, and intestinal epithelial cells. These macrophages also modulate T cell activities and functions, via the secretion of IL-10 for Tregs and IL-1β for Th17 cells. In addition, they affect ILC3 cells through the production of IL-1α and IL-1β. The migration of antigen-loaded CX3CR1high intestinal macrophages to mesenteric lymph nodes is impaired by intestinal microbiota, thus affecting antigen presentation to T cells and effectively sustaining tolerance towards commensal bacteria. On the other hand, XCR1+ DCs play a tolerogenic role upon recognizing commensal bacterial components, while CD103+ cDC2s seem to be important for initiating oral tolerance through their capacity to generate RA and IL-10. In IBD, large numbers of Ly6Chigh inflammatory monocytes are recruited to the intestine in a CCR2-dependent manner, becoming pro-inflammatory effector cells. These inflammatory macrophages produce TNFα, IL-6, and iNOS, and directly cause the onset and development of fibrosis through a disproportionate accumulation of ECM. The intestinal microbiota is impaired during chronic colitis, and CX3CR1high macrophages can change their habits and migrate to lymph nodes. CD103+ cDC numbers are significantly reduced in the inflamed and uninflamed intestine in IBD; however, activated DCs can release inflammatory cytokines, in addition to type I IFN produced by pDCs. All these phenomena contribute to a generalized inflammation. Part of the figure was generated by using pictures from Servier Medical Art.
Figure 2. Macrophages and dendritic cell in homeostasis and IBD. Macrophages and DCs play important roles in homeostasis and in the development of IBD by phagocytosing cellular debris, producing cytokines, regulating tissue repair, and interacting with other cells. In the intestine, monocyte-derived macrophages (MΦ) are more abundant than tissue-resident macrophages of embryonic origin. Both perform phagocytosis, produce cytokines, and interact with other cells. Upon weaning, bone marrow-derived monocytes egress from the circulation and extravasate into the tissue, where they undergo differentiation and maturation (downregulation of Ly6C, production of MHCII, and increased expression of CX3CR1). In homeostasis, tissue-resident macrophages in the muscularis externa interact with enteric and myenteric neurons controlling intestinal secretion and motility, while in the lamina propria, macrophages provide signals to intestinal stem cells that give rise to goblet cells, Paneth cells, and intestinal epithelial cells. These macrophages also modulate T cell activities and functions, via the secretion of IL-10 for Tregs and IL-1β for Th17 cells. In addition, they affect ILC3 cells through the production of IL-1α and IL-1β. The migration of antigen-loaded CX3CR1high intestinal macrophages to mesenteric lymph nodes is impaired by intestinal microbiota, thus affecting antigen presentation to T cells and effectively sustaining tolerance towards commensal bacteria. On the other hand, XCR1+ DCs play a tolerogenic role upon recognizing commensal bacterial components, while CD103+ cDC2s seem to be important for initiating oral tolerance through their capacity to generate RA and IL-10. In IBD, large numbers of Ly6Chigh inflammatory monocytes are recruited to the intestine in a CCR2-dependent manner, becoming pro-inflammatory effector cells. These inflammatory macrophages produce TNFα, IL-6, and iNOS, and directly cause the onset and development of fibrosis through a disproportionate accumulation of ECM. The intestinal microbiota is impaired during chronic colitis, and CX3CR1high macrophages can change their habits and migrate to lymph nodes. CD103+ cDC numbers are significantly reduced in the inflamed and uninflamed intestine in IBD; however, activated DCs can release inflammatory cytokines, in addition to type I IFN produced by pDCs. All these phenomena contribute to a generalized inflammation. Part of the figure was generated by using pictures from Servier Medical Art.
Ijms 24 01526 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Saez, A.; Herrero-Fernandez, B.; Gomez-Bris, R.; Sánchez-Martinez, H.; Gonzalez-Granado, J.M. Pathophysiology of Inflammatory Bowel Disease: Innate Immune System. Int. J. Mol. Sci. 2023, 24, 1526. https://doi.org/10.3390/ijms24021526

AMA Style

Saez A, Herrero-Fernandez B, Gomez-Bris R, Sánchez-Martinez H, Gonzalez-Granado JM. Pathophysiology of Inflammatory Bowel Disease: Innate Immune System. International Journal of Molecular Sciences. 2023; 24(2):1526. https://doi.org/10.3390/ijms24021526

Chicago/Turabian Style

Saez, Angela, Beatriz Herrero-Fernandez, Raquel Gomez-Bris, Hector Sánchez-Martinez, and Jose M. Gonzalez-Granado. 2023. "Pathophysiology of Inflammatory Bowel Disease: Innate Immune System" International Journal of Molecular Sciences 24, no. 2: 1526. https://doi.org/10.3390/ijms24021526

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop