Fact-checked by Grok 2 weeks ago

Phagocyte

Phagocytes are specialized that perform , a fundamental cellular process involving the recognition, engulfment, and destruction of large particles greater than 0.5 μm in diameter, such as microorganisms, foreign substances, and apoptotic cells. These cells form a critical component of the , rapidly clearing pathogens and debris to prevent and maintain , while also contributing to adaptive immunity through . Discovered by in the late 19th century through observations of larvae, phagocytosis was recognized as a key immunobiological mechanism, earning Metchnikoff the in or in 1908. Phagocytes are broadly classified into professional and non-professional types based on their efficiency in particle ingestion. Professional phagocytes, which exhibit high phagocytic activity, include neutrophils, monocytes, macrophages, dendritic cells, , and osteoclasts; these cells express a variety of receptors, such as Fcγ receptors and complement receptors, to recognize opsonized targets and initiate engulfment via remodeling. Neutrophils, the most abundant circulating leukocytes (approximately 4.2 × 10⁹/L in humans), provide rapid responses to bacterial infections through , release of , and formation of (NETs), though they have a short lifespan of approximately 5 days in circulation. Macrophages, often tissue-resident and long-lived (e.g., up to 4.2 years for in the ), serve as sentinels for chronic surveillance, clearing apoptotic cells and orchestrating resolution. Monocytes, their circulating precursors, differentiate into macrophages or dendritic cells upon entering tissues, supporting immune surveillance with a circulation of around 20 hours. Dendritic cells, marrow-derived and short-lived, with a circulating lifespan of approximately 1–2 days, excel in to T cells, bridging innate and adaptive responses. In addition to pathogen elimination, phagocytes regulate immune tolerance by efficiently removing billions of apoptotic cells daily without triggering inflammation, a process essential for preventing autoimmunity and promoting tissue repair. Defects in phagocyte function, such as those seen in chronic granulomatous disease, compromise host defense and lead to recurrent infections, underscoring their indispensable role in immunity.

Fundamentals

Definition and Characteristics

Phagocytes are specialized immune cells that engulf and destroy pathogens, cellular debris, and apoptotic cells through the process of , a fundamental mechanism in innate immunity and . represents a specialized form of dedicated to internalizing large particles exceeding 0.5 μm in diameter, distinguishing it from other endocytic pathways that handle smaller entities. This capability enables phagocytes to clear microbial invaders and maintain cellular integrity by removing damaged or dying cells. Structurally, phagocytes are equipped with key organelles and components essential for their function, including phagosomes—membrane-derived vesicles that encapsulate engulfed material—and lysosomes that fuse with phagosomes to form phagolysosomes, creating an acidic environment ( as low as 4.5) for enzymatic . The cytoskeleton plays a critical role, with actin polymerization driven by complexes like Arp2/3 and regulated by (such as Rac, Cdc42, and RhoA) facilitating pseudopod extension and particle . Phagocytes are broadly classified into professional and non-professional types based on phagocytic efficiency: professional phagocytes, including macrophages, neutrophils, monocytes, dendritic cells, and , execute phagocytosis as a primary with high proficiency, whereas non-professional phagocytes, such as fibroblasts, epithelial cells, and endothelial cells, perform it less efficiently, primarily for apoptotic cell clearance. Morphologically, phagocytes exhibit features adapted to their roles, with macrophages typically being large cells (20–30 μm in diameter) featuring irregular or kidney-shaped nuclei and abundant, vacuole-rich that supports extensive phagocytic activity. In contrast, neutrophils display multilobulated nuclei and granular containing enzymes and granules, enabling rapid responses to despite their smaller size (around 10–12 μm). These traits underscore the diversity among phagocyte subtypes while unifying their core engulfment and destructive capabilities.

Biological Roles and Importance

Phagocytes constitute the primary effectors of innate immunity, acting as the first line of defense against invading pathogens by rapidly recognizing, engulfing, and destroying microbial threats through . This process limits dissemination and initiates inflammatory responses to additional immune cells, ensuring in the absence of prior exposure. Beyond pathogen control, phagocytes play crucial roles in homeostasis by clearing apoptotic cells and , a process known as that prevents the release of intracellular contents and subsequent . In humans, phagocytes process approximately 10 to 100 billion apoptotic cells daily, primarily neutrophils from circulation, maintaining and avoiding from uncleared . Additionally, phagocytes contribute to tissue remodeling during embryonic development and , where they prune unnecessary structures and orchestrate reorganization to support regeneration and resolution. In healthy states, they sustain sterile environments in barrier tissues such as the lungs and gut; alveolar macrophages in the lungs surveil for airborne while minimizing , and intestinal macrophages balance tolerance with rapid clearance of invasive microbes to preserve mucosal integrity. Phagocytes also bridge innate and adaptive immunity by processing engulfed antigens and presenting them via molecules to T cells, thereby priming specific humoral and cellular responses. In , phagocyte dysregulation drives chronic inflammation, as seen in where monocyte-derived macrophages accumulate oxidized in arterial walls, perpetuating plaque formation and vascular damage. In cancer, tumor-associated macrophages contribute to immune surveillance by phagocytosing malignant cells expressing "eat me" signals, although their often shifts to pro-tumorigenic states that suppress anti-tumor immunity. Recent post-2020 studies highlight phagocytic roles in , particularly in , where impaired of amyloid-beta aggregates and synaptic debris exacerbates plaque buildup and neuronal loss due to genetic risk factors like TREM2 variants; as of 2025, research has further shown that accumulation in impairs , and their clearance enhances Aβ uptake.

Historical Development

Early Observations

The earliest observations of white blood cell mobility in tissues were made by German pathologist Julius Cohnheim in 1867, who demonstrated through microscopic studies of inflamed frog mesentery that leukocytes could migrate across intact capillary walls via amoeboid movement, a process he termed diapedesis. This finding established the foundational concept of leukocyte emigration from blood vessels to sites of injury or infection, laying the groundwork for understanding cellular involvement in inflammation. A pivotal advancement came in 1882 when Russian zoologist , while studying embryonic development in larvae at the Messina Marine Biological Station, observed mobile mesodermal cells actively engulfing foreign particles. In a seminal experiment, Metchnikoff inserted rose thorns into the transparent larvae and noted that phagocytic cells rapidly surrounded and internalized the intruding material, interpreting this as a defensive cellular response against nonself entities. This discovery introduced the term "" to describe the process, shifting focus from passive humoral defenses to active cellular immunity. Metchnikoff extended these insights through experiments on bacterial infections, particularly using bacilli () in guinea pigs during the 1880s. He injected the bacilli subcutaneously and observed that mobile leukocytes at the site of injection engulfed and destroyed the pathogens in the exuded fluid, with similar phagocytic activity noted in the . These studies, conducted at the , highlighted the protective role of phagocytes in resisting , a model for understanding cellular defenses against microbial threats. These early findings sparked intense debate within the scientific community regarding the primacy of cellular versus . Metchnikoff championed as the central defensive process, but this view clashed with the toxin-based humoral theories advocated by bacteriologists like , who emphasized soluble factors and antitoxins as the main agents of protection against diseases such as . The controversy underscored initial misconceptions that cellular activity was merely secondary to chemical defenses, delaying widespread of phagocytes' independent role until later experimental validations.

Key Milestones and Researchers

In 1908, and were awarded the in Physiology or Medicine for their foundational work on immunity, with Metchnikoff's contributions emphasizing the role of phagocytes in cellular immunity through his earlier observations of in larvae and mammals. Metchnikoff, often regarded as the father of cellular immunology, demonstrated in the late 19th and early 20th centuries that phagocytes actively engulf and destroy pathogens, laying the groundwork for understanding innate immunity. The mid-20th century brought advances in visualizing phagocyte function through electron microscopy, notably in the when studies revealed the of phagosomes with lysosomes, elucidating the intracellular digestion process essential for elimination. This technological leap shifted research from light microscopy to ultrastructural details, enabling precise mapping of phagocytic compartments. By the , the identification of (ROS) as key antimicrobial agents in phagocytes marked a pivotal biochemical milestone, with Bernard Babior's work at the Scripps Clinic demonstrating that activated neutrophils generate via the complex during the respiratory burst. This discovery explained the molecular basis of oxidative killing and linked defects in ROS production to . The transition to in the late 20th and early 21st centuries accelerated with genetic tools; for instance, the saw CRISPR-Cas9 applications in dissecting phagocytic receptors, including studies on Fcγ receptors that mediate antibody-dependent in macrophages. Steinman, who co-discovered dendritic cells in the 1970s and received the 2011 Nobel Prize in Physiology or Medicine for this work, highlighted their unique phagocytic and antigen-presenting roles, bridging innate and adaptive immunity. Recent advances, such as single-cell sequencing since the mid-2010s, have unveiled the heterogeneity of phagocyte populations, revealing diverse transcriptional states in macrophages across tissues and contexts. This evolution from morphological observations to genomic and functional profiling underscores the field's progression toward precision .

Phagocytosis Mechanism

Recognition and Engulfment

Phagocytes recognize and bind to target particles, such as pathogens or apoptotic cells, through specialized surface receptors that initiate the engulfment process. This recognition phase is crucial for distinguishing harmful entities from host tissues and ensuring selective internalization. Recognition can occur via opsonin-dependent or non-opsonic mechanisms, each involving distinct receptor-ligand interactions. In opsonin-dependent recognition, soluble host proteins known as opsonins coat the target, enhancing its visibility to phagocytes. Antibodies, particularly (IgG), bind to microbial antigens and are detected by Fcγ receptors (FcγR) on the phagocyte surface, triggering signaling cascades that promote attachment and uptake. Complement proteins, such as C3b, opsonize targets following activation of the and are recognized primarily by (CR1) and complement receptor 3 (CR3), facilitating phagocytosis of and immune complexes. These opsonic pathways amplify the efficiency of recognition, particularly for immune complexes and opsonized pathogens. Non-opsonic recognition allows direct binding without opsonin intermediaries, relying on receptors that detect specific molecular motifs. For instance, the on macrophages binds to mannose-containing glycans on fungal and bacterial surfaces, enabling uptake of pathogens like species. In the clearance of apoptotic cells, exposed on the outer membrane leaflet is recognized by receptors such as stabilin-2 or TIM-4, promoting non-inflammatory engulfment to maintain . These mechanisms ensure of unopsonized targets, including certain microbes and dying cells. Upon recognition, engulfment proceeds through dynamic cytoskeletal rearrangements that internalize the target into a membrane-bound phagosome. The process begins with receptor clustering at the contact site, leading to localized actin polymerization that drives pseudopod extension around the particle, forming a phagocytic cup. Actin filaments, nucleated by the Arp2/3 complex, provide the protrusive force necessary for membrane protrusion and target enclosure. The phagocytic cup seals at the base, pinching off to generate an intracellular phagosome containing the engulfed material. Engulfment mechanisms vary based on target size and receptor type, with two primary modes observed: zippering and sinking. Zippering phagocytosis, typically mediated by Fcγ receptors, involves sequential engagement of receptors along the target surface, resulting in tight pseudopod wrapping and efficient uptake of small to medium particles like opsonized . In contrast, sinking phagocytosis, often driven by complement receptors like CR3, submerges the target into the cell interior without extensive pseudopod formation, suitable for larger or deformable particles such as erythrocytes. These modes ensure adaptability to diverse targets while maintaining integrity. The engulfment process is energy-intensive and ATP-dependent, relying on to fuel dynamics and membrane remodeling. Rho family , including Rac1 and Cdc42, play pivotal roles in orchestrating cytoskeletal changes; Rac1 promotes lamellipodia-like pseudopod extension, while Cdc42 drives formation and initial nucleation at the phagocytic site. These activate downstream effectors like and N-WASP to stimulate Arp2/3-mediated branching of actin filaments, ensuring coordinated pseudopod progression and closure. Efficiency of recognition and engulfment varies among phagocyte types, influenced by receptor expression and cytoskeletal capacity. Neutrophils exhibit high phagocytic rates for small pathogens, capable of engulfing up to 50 per cell, whereas macrophages handle larger particles more effectively due to their expansive pseudopods. Dendritic cells, while proficient in antigen sampling, show moderated engulfment to preserve target integrity for . Monocytes display lower baseline efficiency, which increases upon into macrophages.

Intracellular Killing and Digestion

Following the closure of the during engulfment, phagocytes initiate a series of maturation events that transform the nascent into a microbicidal compartment capable of destroying engulfed pathogens. The progressively fuses with early endosomes, late endosomes, and ultimately lysosomes, culminating in the formation of the phagolysosome. This maturation process involves the recruitment of Rab GTPases and SNARE proteins, which facilitate fusion and the delivery of lysosomal contents. A critical aspect of phagolysosome function is acidification, primarily driven by the vacuolar-type H+-ATPase () , which lowers the intraluminal from neutral to approximately 4.5–5.5. This acidic environment activates lysosomal hydrolases and enhances the potency of antimicrobial agents, creating conditions hostile to microbial survival. Within the phagolysosome, killing occurs through both oxygen-dependent and oxygen-independent mechanisms. The oxygen-dependent pathway, prominent in neutrophils and macrophages, relies on the phagocyte complex (), which assembles upon phagosome activation to generate anion (O2•−) from molecular oxygen and NADPH. is rapidly converted to (H2O2), which (MPO) then uses to produce (HOCl), a potent oxidant that damages microbial proteins, , and DNA. This process, known as the respiratory burst, dramatically increases oxygen consumption and is essential for efficient killing of catalase-negative . Complementing the oxidative burst, oxygen-independent mechanisms involve granule-derived antimicrobial proteins and enzymes released into the phagolysosome. These include , which hydrolyzes bacterial ; , small cationic peptides that disrupt microbial membranes; cationic proteins like bactericidal/permeability-increasing protein (BPI); and , which sequesters iron to starve pathogens. These agents provide robust killing even in anaerobic conditions or in phagocytes with impaired oxidative function, such as those in . Once killing is achieved, digestion proceeds via hydrolytic enzymes within the phagolysosome, breaking down microbial remnants into soluble components for . Key enzymes include cathepsins (aspartic and cysteine proteases that degrade proteins), DNases (which fragment nucleic acids), and other hydrolases such as lipases and glycosidases. This enzymatic degradation not only eliminates threats but also allows phagocytes to extract , , and for their own metabolic needs and . The efficiency of intracellular killing varies by phagocyte type; for instance, neutrophils exhibit a particularly intense respiratory burst, enabling rapid destruction of engulfed particles within minutes. While the focus here is on intracellular processes, neutrophils can also deploy (NETs) as an alternative extracellular killing strategy, involving the release of and proteins.

Phagocyte Activation and Mobility

Activation Pathways

Phagocytes are primed for activation through various exogenous and endogenous signals that enhance their functional readiness. Cytokines such as interferon-gamma (IFN-γ) serve as key priming agents, binding to receptors on phagocytes to initiate intracellular signaling that promotes pro-inflammatory states, particularly in macrophages. Pathogen-associated molecular patterns (PAMPs), like (LPS) from , bind to Toll-like receptors (TLRs), such as TLR4, triggering rapid immune responses in both macrophages and neutrophils. Similarly, damage-associated molecular patterns (DAMPs), including (HMGB1) and ATP released from stressed or necrotic cells, act as danger signals that engage receptors (PRRs) like TLRs and , amplifying phagocyte alertness without direct . Upon signal recognition, phagocytes activate distinct intracellular pathways that drive transcriptional and metabolic reprogramming. The pathway, activated via TLR signaling or receptors, translocates to the to upregulate pro-inflammatory genes, fostering a cytotoxic in activated phagocytes. In macrophages, IFN-γ engages the JAK-STAT1 axis, promoting classical () polarization by inducing expression of genes involved in antimicrobial defense. Metabolic shifts accompany these pathways; for instance, M1-like activation upregulates and the to support rapid energy demands and biosynthesis, contrasting with favored in alternative () states. Activation outcomes include enhanced expression of surface receptors, such as scavenger receptors and molecules, which improve recognition and . Increased production of (ROS) via NADPH oxidase assembly bolsters intracellular killing capabilities in primed phagocytes. Macrophages exhibit polarization plasticity, with M1 states (pro-inflammatory, antitumor) driven by IFN-γ and LPS, versus M2 states (anti-inflammatory, tissue repair) induced by IL-4 or IL-10. Research indicates this spectrum's adaptability in tumor microenvironments, where metabolic interventions can shift tumor-associated macrophages toward antitumor phenotypes. In neutrophils, activation yields rapid, transient responses—often within seconds—characterized by intense ROS bursts and , differing from the sustained, adaptable activation in macrophages that supports prolonged .

Migration and Chemotaxis

Phagocytes exhibit , the directed migration toward sites of or injury along chemical gradients established by soluble chemoattractants such as complement component C5a, (LTB4), and N-formyl-methionyl-leucyl-phenylalanine (fMLP). These molecules, released by pathogens, damaged cells, or activated immune components, create concentration gradients that guide phagocytes from the bloodstream or surrounding tissues to the target area. The process is mediated by G-protein-coupled receptors (GPCRs) on the phagocyte surface, including formyl peptide receptor 1 (FPR1), which specifically binds formylated peptides like fMLP derived from bacteria. Upon ligand binding, these receptors activate intracellular signaling cascades involving heterotrimeric G-proteins, leading to cytoskeletal polarization essential for motility. Actin polymerization is enriched at the leading edge, forming lamellipodia or filopodia that propel the cell forward, while myosin II contracts at the rear uropod to facilitate retraction and maintain directionality. In tissues, phagocytes navigate complex environments through diapedesis, the transmigration across endothelial barriers, primarily via homophilic interactions between on leukocytes and endothelial cells, which destabilize junctions to allow passage without disrupting the barrier integrity. To penetrate the , phagocytes secrete matrix metalloproteinases (MMPs), such as MT1-MMP, which degrade ECM components like and , creating paths for interstitial migration. Migration speed is tightly regulated, with neutrophils typically moving at 10-20 μm/min in response to gradients, balancing rapid with coordinated tissue traversal. Recent intravital studies from 2022 have revealed swarm behavior in tissues, where lead neutrophils amplify signals to followers, enabling collective and efficient containment of infections through amplified chemotactic relays. This is often triggered following phagocyte , integrating with broader immune responses.

Types of Phagocytes

Monocytes and Macrophages

Monocytes are circulating mononuclear phagocytes that primarily originate from hematopoietic progenitors in the . In mice, these include Ly6C-high monocytes, which serve as precursors for tissue under inflammatory conditions, while in humans, CD14-positive monocytes represent a key subset with similar roles. Upon recruitment to tissues, monocytes differentiate into , which are long-lived, tissue-resident professional phagocytes adapted to specific microenvironments. For instance, in the lungs, they become alveolar that maintain pulmonary , while in the liver, they develop into Kupffer cells responsible for clearing blood-borne pathogens and debris. This differentiation is influenced by local growth factors, such as (GM-CSF) for alveolar . Bone marrow-derived monocytes can also contribute to self-renewing populations in adult tissues when embryonic-derived cells are depleted. Macrophages perform essential functions in long-term immune surveillance, patrolling tissues for pathogens and damaged cells, and engaging in efferocytosis—the phagocytosis of apoptotic cells—to prevent inflammation and promote resolution. They secrete cytokines such as interleukin-1 (IL-1) and tumor necrosis factor-alpha (TNF-α), particularly in pro-inflammatory states, to orchestrate immune responses. Macrophages exhibit functional polarization, adopting M1-like phenotypes for antimicrobial activity and M2-like phenotypes for tissue repair and anti-inflammatory effects, driven by environmental cues. A hallmark of macrophages is their high , allowing phenotypic shifts in response to stimuli, and their capacity for self-renewal within tissues, independent of continuous influx under steady-state conditions. Recent studies from 2023 onward have highlighted epigenetic modifications in monocytes that persist after severe acute respiratory syndrome coronavirus 2 () infection, contributing to dysregulated responses in and other chronic inflammatory diseases. In terms of phagocytic capacity, macrophages efficiently engulf large particles, such as or apoptotic bodies, but operate at a slower rate compared to neutrophils, emphasizing their role in sustained rather than acute clearance.

Neutrophils

Neutrophils are the most abundant type of circulating leukocytes, comprising 50% to 70% of the total white blood cell count in healthy adults. These granulocytes are characterized by a multilobular nucleus, typically segmented into 2 to 5 lobes connected by thin filaments, which facilitates their deformability during migration through tissues. Neutrophils contain distinct granule populations, including azurophilic (primary) granules that store antimicrobial enzymes like myeloperoxidase and defensins, and specific (secondary) granules that hold lactoferrin, lysozyme, and neutrophil gelatinase-associated lipocalin for targeted release during immune responses. As key effectors in acute , neutrophils rapidly respond to sites through and , where they perform to engulf and destroy and fungi. Upon contact, they generate a potent oxidative burst via , producing (ROS) such as and to damage microbial targets, with this process being more intense in neutrophils compared to other phagocytes due to their high enzyme content.00830-8) Additionally, neutrophils release (NETs), web-like structures composed of decondensed chromatin fibers decorated with antimicrobial proteins like histones, , and , which trap and kill extracellular pathogens without requiring engulfment. The lifecycle of neutrophils is short and tightly regulated to prevent excessive . Mature neutrophils have a circulating of approximately 4–18 hours under homeostatic conditions, with total lifespan estimated at up to 5.4 days. In tissues during , their functional activity can persist for several days. Following clearance, activated neutrophils undergo , a process that signals for their recognition and by macrophages, thereby resolving and avoiding tissue damage from prolonged activity. Recent research highlights the dual role of neutrophils in pathology, particularly through dysregulated formation. In autoimmune conditions like systemic lupus erythematosus (SLE), excessive contribute to disease progression by promoting autoantibody production and vascular inflammation, as evidenced by elevated levels correlating with lupus flares in studies from 2021 onward. This underscores neutrophils' involvement in both protective immunity and potential when their mechanisms are unchecked.

Dendritic Cells

Dendritic cells represent a specialized class of professional phagocytes that link innate and adaptive immunity by capturing, processing, and presenting antigens to initiate targeted T-cell responses. Originating from hematopoietic stem cells in the , they differentiate into distinct lineages that patrol peripheral tissues in an immature state, actively surveying for pathogens and damaged cells. Upon , these cells undergo maturation, migrating to lymphoid organs to orchestrate immune priming, thereby distinguishing them from other phagocytes focused primarily on immediate microbial clearance. Dendritic cells encompass several subtypes, including conventional dendritic cells (cDCs) divided into cDC1 and cDC2 subsets, as well as plasmacytoid dendritic cells (pDCs). cDC1s specialize in cross-presenting antigens via to activate ⁺ T cells, while cDC2s primarily present antigens on to stimulate ⁺ T cells; pDCs, in contrast, excel at producing type I interferons in response to viral threats. All subtypes arise from common precursors, such as common dendritic cell progenitors (CDPs), which commit to either cDC or pDC lineages through transcription factors like IRF8 and BATF3 for cDC1s. A core function of dendritic cells involves sampling in peripheral tissues through macropinocytosis, an actin-driven process that enables the non-specific uptake of , soluble proteins, and particulates for subsequent processing. Following engulfment, antigens are degraded in endolysosomal compartments and loaded onto molecules. Activated dendritic cells then migrate to nodes via upregulation of CCR7, a that guides them to T-cell zones, where they present peptide-MHC complexes to prime naive T cells and direct their into effector subsets.00455-X) Immature dendritic cells exhibit exceptionally high endocytic capacity, far surpassing that of other antigen-presenting cells, which facilitates broad environmental surveillance. Maturation is triggered by (TLR) ligation, where pathogen-derived ligands bind TLRs to activate signaling cascades like , leading to profound phenotypic changes. This includes downregulation of endocytic receptors and upregulation of costimulatory molecules such as and , which provide signal 2 to T cells for full activation, alongside increased expression for enhanced display.00449-4) Recent single-cell RNA sequencing studies from 2023 have illuminated heterogeneity, revealing diverse transcriptional states within tumor-infiltrating subsets that influence efficacy and outcomes. For example, analyses in non-small cell lung cancer and identified cDC2 subpopulations with pro- versus profiles, correlating with anti-PD-1 response rates and guiding subset-specific targeting in therapeutic s. These insights underscore the functional diversity of s, enabling tailored strategies to boost antitumor immunity.

Other Professional Phagocytes

In addition to the primary types, other cells exhibit professional phagocytic activity. , granulocytes derived from , perform phagocytosis primarily against parasitic helminths and allergens, releasing cytotoxic granules and producing ROS, though with lower efficiency compared to neutrophils. Osteoclasts, multinucleated cells differentiated from monocyte-macrophage lineage, specialize in by phagocytosing mineralized bone matrix and debris, playing a key role in skeletal remodeling and calcium . These cells express phagocytic receptors like Fcγ and complement receptors, contributing to specialized immune and functions.

Non-Professional Phagocytes

Non-professional phagocytes are tissue-resident cells that engage in opportunistically, primarily to maintain local rather than as a dedicated immune function. These cells, including epithelial cells, fibroblasts, and endothelial cells, exhibit limited phagocytic capacity compared to professional phagocytes like macrophages and neutrophils, which possess specialized machinery for pathogen engulfment and destruction. Epithelial cells in the gut and lungs, for instance, uptake cellular and apoptotic to support barrier , while fibroblasts at wound sites clear remnants, and endothelial cells handle vascular cleanup of senescent red blood cells or microthrombi. cells in the vasculature also act as non-professional phagocytes, engulfing apoptotic neighbors during progression. These activities rely on a restricted set of receptors, such as (e.g., αvβ3) and , which recognize "eat-me" signals like on target cells, but with efficiencies typically below 10-15% , far lower than professional counterparts. Unlike immune-specialized cells, non-professional phagocytes lack robust intracellular killing mechanisms, such as potent lysosomal enzymes or production tailored for microbial destruction, limiting their role to non-infectious . In physiological contexts, these cells contribute to tissue homeostasis by preventing the accumulation of necrotic material that could disrupt organ function, such as in epithelial clearance of surfactant-laden debris or intestinal epithelial to sustain . Pathologically, cell-mediated in atherosclerotic plaques helps mitigate by removing apoptotic cells, reducing necrotic core formation and plaque instability when efficient. Emerging research highlights roles in microbiome interactions, where neonatal intestinal epithelial cells efferocytose Salmonella-infected enterocytes, potentially modulating composition by limiting dissemination and without adult-like shedding mechanisms. However, the limited capacity of non-professional phagocytes makes them susceptible to overload in conditions, where excessive apoptotic burden can lead to incomplete clearance and formation as a compensatory response. In , impaired by these cells due to factors like oxidized lipids or clonal expansion exacerbates plaque vulnerability. Similarly, in microbiome contexts, age-dependent inefficiencies in epithelial may allow transient persistence, underscoring their auxiliary rather than primary defensive role.

Immune Interactions

Role in Apoptosis Clearance

Phagocytes play a crucial role in , the specialized process of recognizing, engulfing, and degrading to maintain without triggering . This clearance is mediated by "eat-me" signals on , such as the externalization of (PS) on the plasma membrane, which binds to receptors like TIM-4 and stabilin-2 on phagocytes to initiate engulfment. Another key signal is the exposure of on the cell surface, which activates the low-density lipoprotein receptor-related protein (LRP) on phagocytes via trans-activation, facilitating rapid uptake. Following engulfment, phagocytes process the in lysosomes, preventing the release of intracellular contents that could provoke immune responses. Efferocytosis also induces anti-inflammatory signaling in phagocytes, promoting the production of immunosuppressive cytokines such as transforming growth factor-β (TGF-β) and interleukin-10 (IL-10), which dampen pro-inflammatory pathways and support resolution of . Macrophages serve as the primary effectors of , efficiently clearing apoptotic cells in tissues, while neutrophils and dendritic cells provide supplementary assistance, particularly in dynamic environments like inflamed sites. In humans, this process clears approximately 200–300 billion apoptotic cells daily, underscoring its scale in preserving . The importance of efferocytosis lies in its ability to prevent secondary necrosis of uncleared apoptotic cells, which would otherwise release damage-associated molecular patterns (DAMPs) and trigger chronic or . Defects in this process are implicated in autoimmune disorders, notably systemic lupus erythematosus (SLE), where impaired clearance leads to accumulation of apoptotic debris and autoantibody production against nuclear antigens. Recent 2022 studies have highlighted efferocytosis's dual role in resolution and cancer; for instance, it promotes reprogramming in resolving tissues, but in tumors, efferocytosis by tumor-associated macrophages (TAMs) can drive M2-like polarization, fostering an immunosuppressive microenvironment that supports tumor growth and . Interventions like thymosin α1 have been shown to reverse this M2 polarization during efferocytosis, enhancing anti-tumor immunity.

Antigen Presentation

Phagocytes, particularly dendritic cells (DCs) and macrophages, play a crucial role in bridging innate and adaptive immunity by processing engulfed antigens and presenting fragments on (MHC) molecules to T lymphocytes. Following , antigens are internalized into phagosomes, where they undergo degradation by lysosomal proteases such as cathepsins, generating peptides suitable for MHC loading. These peptides are then loaded onto molecules in endosomal compartments for presentation to + T cells, or, in the case of , onto molecules for CD8+ T cell activation. In presentation, newly synthesized molecules associate with the invariant chain (, also known as CD74) in the to prevent premature binding and direct the complex to phagosomal or -rich compartments (MIIC). There, is proteolytically degraded, leaving a CLIP fragment that is exchanged for antigenic with the aid of , enabling stable - complexes to traffic to the cell surface. For presentation of exogenous phagosomal antigens, is predominant in DCs, where antigens escape to the for proteasomal degradation, and resulting are transported via the transporter associated with antigen processing () into the or phagosomes for loading onto recycled molecules. This process often involves ER-phagosome fusion mediated by proteins like Sec22b, enhancing efficiency in professional antigen-presenting cells. Dendritic cells are the primary phagocytes specialized for , particularly by the cDC1 subset, which efficiently primes naive + T cells against intracellular pathogens and tumors. Macrophages serve a secondary role, capable of via vacuolar or cytosolic pathways but generally less effective at priming naive T cells compared to DCs, often supporting reactivation of T cells or antigen transfer to DCs. TAP recruitment to phagosomes in DCs further boosts MHC I supply, while the invariant chain ensures targeted MHC II loading, optimizing overall presentation efficiency. Effective T cell activation requires not only MHC-peptide display but also costimulatory signals, such as the interaction between B7 molecules (CD80 and CD86) on phagocytes and CD28 on T cells, which promotes T cell proliferation and survival. DCs upregulate B7 upon maturation, providing robust costimulation, whereas macrophages offer variable levels depending on their activation state. The cytokine environment further shapes responses; for instance, IL-12 secreted by DCs during antigen presentation drives Th1 differentiation and IFN-γ production by CD8+ T cells, enhancing cytotoxic immunity. Recent advances, including cryo-electron microscopy (cryo-EM) structures post-2018, have illuminated the in atomic detail, revealing how tapasin and associated chaperones like ERp57 and edit peptides for high-affinity binding in professional APCs such as DCs. These insights, derived from 9.9-Å cryo-EM densities combined with simulations, demonstrate the 's layered architecture and glycan-mediated stabilization, underscoring mechanisms that fine-tune efficiency in phagocytes.

Contribution to Immunological Tolerance

Phagocytes play a crucial role in maintaining immunological tolerance by suppressing aberrant immune responses against self-antigens and harmless environmental antigens, thereby preventing autoimmunity and chronic inflammation. Regulatory dendritic cells (DCs) contribute to this process by presenting self-antigens in the context of low costimulatory signals, such as reduced expression of CD80 and CD86, which promotes T cell anergy or deletion rather than activation. Similarly, macrophages secrete immunosuppressive cytokines like interleukin-10 (IL-10) and transforming growth factor-β (TGF-β), which dampen effector T cell responses and foster regulatory T cell (Treg) expansion to enforce peripheral tolerance. These mechanisms also facilitate the peripheral deletion of autoreactive T cells through phagocyte-mediated apoptosis induction or cross-presentation of self-antigens in a tolerogenic manner. Specific subtypes of phagocytes are specialized for tolerance induction. Tolerogenic DCs, particularly the CD103+ subset in mucosal tissues, excel at capturing and presenting antigens from apoptotic cells or commensals while maintaining an immature phenotype that limits costimulation and promotes Treg differentiation via retinoic acid production. M2-polarized macrophages, characterized by their anti-inflammatory profile, further support tolerance by enhancing IL-10 and TGF-β secretion in response to steady-state signals, thereby resolving inflammation and maintaining tissue homeostasis without excessive immune activation. In physiological contexts, phagocytes uphold tolerance through steady-state —the efficient clearance of apoptotic cells—which prevents the release of pro-inflammatory damage-associated molecular patterns and sustains an milieu via IL-10 and TGF-β upregulation. In the gut, mononuclear phagocytes, including CD103+ DCs and resident macrophages, interact with commensal to induce tolerance, sampling luminal antigens and driving + Treg generation to avert responses against the . Defects in these phagocyte functions disrupt tolerance, contributing to diseases such as (IBD), where impaired and dysregulated production lead to chronic mucosal inflammation, and allergies, arising from failed suppression of Th2 responses to environmental antigens. Recent advances highlight the therapeutic potential of phagocyte-derived exosomes in induction. Studies from 2024 demonstrate that exosomes loaded with tolerogenic signals, such as TGF-β, promote Treg expansion and suppress allograft rejection in transplant models, offering a non-cellular approach to achieve long-term without broad . Similarly, M2 macrophage-derived exosomes have been engineered to deliver miRNA cargos that modulate T cell reactivity, enhancing antigen-specific in autoimmune and transplant settings.

Pathogen Evasion Strategies

Avoiding Initial Contact

Pathogens have evolved multiple strategies to evade initial detection and approach by phagocytes, thereby delaying the onset of phagocytosis and allowing time for replication or dissemination. These pre-contact evasion tactics primarily target pattern recognition receptors (PRRs) on phagocytes, such as Toll-like receptor 4 (TLR4), and disrupt signaling pathways that mediate chemotaxis and recruitment. By masking pathogen-associated molecular patterns (PAMPs) or mimicking host structures, microbes prevent the activation of innate immune responses that would otherwise draw phagocytes to the infection site. One prominent tactic is the production of polysaccharide capsules, which physically shield surface PAMPs from recognition by phagocyte receptors and complement proteins. For instance, the thick capsule of sterically hinders the deposition of C3b opsonins on the bacterial surface, thereby inhibiting binding to complement receptors on neutrophils and macrophages. This capsule not only reduces opsonophagocytosis but also limits the exposure of lipoteichoic acid and other PAMPs, delaying phagocyte activation. Similarly, capsules in other pathogens like serve as a non-immunogenic barrier that impairs initial contact. Biofilm formation represents another key strategy, particularly in chronic infections, where extracellular polymeric substances create a protective matrix that sequesters bacteria from circulating phagocytes. In the lungs, Pseudomonas aeruginosa forms biofilms during cystic fibrosis exacerbations, which attenuate neutrophil chemotaxis and phagocytosis by limiting oxygen availability and releasing quorum-sensing molecules that suppress immune signaling. These biofilms embed bacteria in a viscous environment that physically impedes phagocyte penetration and reduces PAMP exposure, allowing persistent colonization despite robust innate responses. Molecular mimicry further enables pathogens to blend with host tissues, avoiding PRR-mediated detection. Bacterial proteins or glycans that structurally resemble host molecules can inhibit complement activation and phagocyte recognition; for example, certain streptococcal surface proteins mimic eukaryotic components, reducing opsonization and chemotactic signals. This mimicry exploits host tolerance mechanisms, preventing the inflammatory cues that recruit monocytes and neutrophils. Pathogens also actively disrupt phagocyte migration through mechanisms that inhibit signals. Many bacteria secrete factors that interfere with G-protein-coupled receptor (GPCR) pathways, such as or formyl peptide receptors, thereby blocking the directional movement of neutrophils toward infection sites. For example, produces chemotaxis-inhibitory proteins that bind and neutralize host chemoattractants, reducing phagocyte accumulation at early infection stages. Complementing this, some pathogens shed decoy antigens or surface components to distract phagocytes, diverting their attention to soluble mimics rather than the viable bacteria. These decoys, often released as vesicles or fragments, bind complement or antibodies, exhausting local immune resources before phagocytes reach the pathogen core. A well-documented example is , the causative agent of , which modifies its (LPS) structure during mammalian infection to evade TLR4 recognition. At 37°C, Y. pestis produces tetra-acylated , a hypoacylated form that binds poorly to the TLR4/MD-2 complex on macrophages and dendritic cells, suppressing proinflammatory production and subsequent phagocyte recruitment. This LPS alteration allows the bacterium to disseminate systemically with minimal initial immune interference, underscoring the evolutionary pressure on pathogens to fine-tune surface molecules for stealth. Collectively, these evasion strategies significantly delay phagocyte recruitment, enabling pathogens to establish footholds in tissues. By impeding the rapid influx of neutrophils and macrophages, microbes prolong the for replication, often leading to overwhelming infections if adaptive immunity is not swiftly engaged.

Resistance to Engulfment

Pathogens employ various strategies to resist engulfment by phagocytes once initial contact has been established, thereby preventing and subsequent . These mechanisms target the cytoskeleton dynamics, phagocytic receptor engagement, and formation processes essential for particle uptake. By interfering with these steps, pathogens can evade capture and persist in the host environment. One prominent strategy involves the secretion of anti-phagocytic surface factors that recruit host complement regulators to inhibit opsonization and receptor-mediated uptake. For instance, the M protein on binds complement , a negative regulator of the , thereby reducing C3b deposition and subsequent complement receptor engagement on phagocytes. This interaction occurs via the central conserved C-repeat region of M protein, enhancing bacterial survival in non-immune human blood. Similarly, the fibrinogen-binding M1 protein of group A Streptococcus sequesters host fibrinogen, preventing its role in opsonization and further impeding activation during attempted engulfment. Pathogens also disrupt cytoskeletal rearrangements critical for pseudopod extension and phagosome closure. In , the effector SopE acts as a (GEF) for Rho such as Cdc42 and Rac1, initially promoting to facilitate bacterial of non-phagocytic cells; however, coordinated with the GTPase-activating protein SptP subsequently deactivates these pathways, limiting full phagocytic in professional phagocytes and allowing partial escape from engulfment. This manipulation exploits the host's actin regulatory network to subvert complete internalization. Receptor interference represents another key tactic, where pathogens directly block phagocytic receptors to halt signaling cascades leading to engulfment. produces , which binds the Fc region of IgG antibodies, sterically hindering interaction with Fcγ receptors on phagocytes and thereby inhibiting opsonin-dependent uptake. This binding prevents the conformational changes in IgG necessary for effective receptor crosslinking and actin recruitment. Complement receptors can similarly be targeted; for example, capsular polysaccharides of bacteria like mask surface epitopes, reducing C3b opsonization and /3 engagement. Specific examples illustrate how pathogens arrest phagosome formation mid-process. Mycobacterium tuberculosis employs multiple effectors, including the protein tyrosine phosphatase PtpA, to interfere with host Rab GTPases and prevent phagosome-lysosome fusion precursors during early engulfment, effectively stalling closure and maturation from within the forming vacuole. This allows the bacterium to reside in an immature phagosomal compartment resistant to acidification. Viruses have evolved analogous evasion tactics, often through accessory proteins that alter surfaces or phagocyte function post-contact. The HIV-1 Nef protein inhibits in infected macrophages by disrupting the focal delivery of recycling endosomes to the phagocytic cup, thereby impairing and pseudopod progression around viral particles or infected debris. This Nef-dependent mechanism reduces the efficiency of engulfment by up to 70% in primary macrophages. These resistance strategies often arise through evolutionary adaptations, including (HGT) of anti-phagocytic loci among bacterial pathogens. In , HGT contributes to variability in genes encoding M-like proteins such as PrtF1, which bind plasminogen and inhibit complement activation, facilitating rapid dissemination of resistance traits across strains. Such transfers, mediated by phages or plasmids, underscore the dynamic evolution of phagocyte evasion in microbial communities.

Survival and Counterattack Inside Phagocytes

Once internalized within phagocytes, pathogens deploy diverse strategies to evade degradation by blocking phagosome maturation and neutralizing antimicrobial defenses. A key survival mechanism is the inhibition of phagolysosome fusion, which deprives the phagosome of hydrolytic enzymes and low pH required for pathogen destruction. Legionella pneumophila, for example, utilizes its Dot/Icm type IV secretion system to inject over 300 effector proteins, including Sid family members like SidC and SidF, that remodel host membrane trafficking pathways and prevent fusion with lysosomes, thereby creating a replicative niche known as the Legionella-containing vacuole (LCV). This system is essential for intracellular replication, as mutants defective in Dot/Icm effectors exhibit restored phagolysosomal fusion and reduced bacterial viability. Pathogens also counteract the phagocyte's oxidative burst by scavenging reactive oxygen species (ROS). Salmonella enterica serovar Typhimurium expresses periplasmic superoxide dismutases SodCI and SodCII, which convert superoxide radicals into less toxic hydrogen peroxide, enabling bacterial survival against NADPH oxidase-derived ROS in the phagosome. These enzymes are critical during infection, as their absence significantly impairs S. Typhimurium persistence in macrophages and reduces virulence in vivo. To actively counterattack, certain pathogens release toxins that compromise phagosomal integrity and promote escape to the . Listeria monocytogenes secretes listeriolysin O (), a pH-sensitive pore-forming cytolysin that oligomerizes in the acidic phagosomal membrane to create pores, allowing cytosolic release and subsequent actin-based motility for cell-to-cell spread. Recent structural analyses have identified specific residues in LLO that enhance pore stability and escape efficiency, underscoring its role in pathogenicity. LLO activity is tightly regulated to avoid premature host cell , peaking at phagosomal levels around 5.5–6.0. Induction of host cell apoptosis represents another counterattack strategy, disrupting phagocyte function and potentially facilitating pathogen dissemination. Mycobacterium tuberculosis triggers caspase-3-dependent in alveolar macrophages, which may limit bacterial spread by containing infected cells while promoting to T cells. Similarly, L. pneumophila via Dot/Icm effectors induces early in macrophages, aiding evasion of innate immunity. Protozoan pathogens like spp. exemplify long-term intracellular persistence by modifying the (PV), a specialized that supports replication. Leishmania amazonensis sabotages host SUMOylation pathways within the PV, suppressing innate immune signaling and preventing full lysosomal fusion, thus maintaining a nutrient-rich, non-degradative environment. This adaptation allows chronic infection, with PV pH stabilized near neutrality to favor parasite survival. Advancements in genetic screening have illuminated host-pathogen dynamics in phagosomal escape. A 2024 study using CRISPR/Cas9 knockout in bone marrow-derived macrophages demonstrated that PDCD6 negatively regulates LC3-associated phagocytosis during L. monocytogenes infection, with its depletion enhancing ROS production, trapping more in phagosomes, and reducing cytosolic replication. Such screens reveal lactate metabolism's role in modulating phagosomal defenses, offering insights into therapeutic targeting. For persistent infections, pathogens like M. tuberculosis enter latency within granulomatous structures, where dormant reside in modified phagocytes, resisting sterilizing immunity through downregulated metabolism and immune evasion. Granulomas encapsulate in foamy macrophages, enabling decades-long persistence without active replication.

Additional Aspects

Phagocyte-Induced Host Damage

Phagocytes, particularly neutrophils and macrophages, can inflict significant collateral damage to host tissues through the overproduction of (ROS) and proteases during inflammatory responses. The complex in phagocytes generates ROS to kill pathogens, but excessive release can oxidize lipids, proteins, and DNA in surrounding cells, leading to and tissue necrosis. Similarly, proteases such as neutrophil elastase are secreted to degrade bacterial components, yet unchecked activity degrades proteins like and , contributing to structural damage in organs such as the lungs. In (COPD), including , neutrophil elastase from activated phagocytes exacerbates alveolar destruction by overwhelming antiprotease defenses like . In acute settings like , dysregulated phagocytes drive storms, where macrophages and neutrophils release excessive pro-inflammatory s such as TNF-α, IL-1β, and IL-6, amplifying and causing multi-organ failure. This hyperactivation leads to vascular leakage, , and , with phagocyte-derived mediators directly contributing to endothelial injury. Chronic inflammatory conditions, such as (RA), feature persistent synovial infiltration by macrophages that sustain inflammation through ROS and production, eroding and bone via matrix metalloproteinases and activation. Post-ischemic further exemplifies this, as recruited neutrophils produce ROS upon reoxygenation, exacerbating myocardial or cerebral damage through and impaired microvascular perfusion. Genetic dysregulation of phagocyte function, as seen in (CGD), arises from deficiencies that impair ROS production for pathogen killing, resulting in persistent infections and formation from uncontrolled inflammation. In CGD, defective generation leads to granulomatous damage in lungs, liver, and . Therapeutic strategies target these pathways; for instance, anti-TNF agents like reduce macrophage-driven inflammation in RA by inhibiting TNF signaling, thereby limiting amplification and joint destruction. As of 2024, research has identified persistent immune dysregulation in , including activation of myeloid cells such as macrophages, contributing to sustained low-grade inflammation and production, with links to symptoms including fatigue and cardiopulmonary sequelae months post-infection.

Evolutionary Origins

Phagocytosis emerged as a defining feature of eukaryotic cells, predating the evolution of adaptive immunity by billions of years and appearing in the last eukaryotic common ancestor (LECA), estimated to have lived between 2.0 and 1.0 billion years ago. This process, involving the engulfment of particulate matter through plasma membrane invaginations, is absent in prokaryotes and is considered a hallmark of eukaryogenesis. Fossil evidence supports its ancient origins, with indirect signs of eukaryotic predation dating to approximately 1.05 billion years ago in red algae like Bangiomorpha pubescens, and more direct traces of eukaryovory from 1.15 to 0.9 billion years ago in the Shaler Supergroup. In single-celled eukaryotes such as amoebae, phagocytosis facilitated nutrient acquisition through bacterivory, with the process likely present in early protists around 2 billion years ago. Closely tied to endosymbiosis, phagocytosis is hypothesized to have enabled the acquisition of mitochondria by an archaeal host, though debates persist on whether it preceded or followed this event; Asgard archaea, potential pre-mitochondrial hosts, lack full phagocytic capability but may have used membrane protrusions for symbiont capture. The core machinery of phagocytosis exhibits remarkable conservation across eukaryotes, from protists to mammals, underscoring its fundamental role in cellular function. Key components, including actin and the Arp2/3 complex for cytoskeletal remodeling, as well as dynamin for membrane fission, trace back to the LECA and are shared widely, enabling pseudopod extension and particle internalization in diverse lineages. Actin homologs in certain archaea, such as Crenarchaeota, show monophyly with eukaryotic actins, suggesting an even deeper prokaryotic ancestry, while Rho family GTPases—crucial regulators of actin dynamics in phagocytosis—are of bacterial origin, likely acquired through horizontal gene transfer. Phagocytic receptors, however, display limited conservation, having evolved independently in various groups from ancient pattern recognition systems that detect microbial signatures, allowing adaptation to specific environmental threats. In metazoans, phagocytosis expanded as a cornerstone of innate immunity, integrating with multicellular defenses to clear pathogens and debris more efficiently than in unicellular ancestors. Comparative studies reveal this elaboration in early-diverging animals; for instance, coelomocytes—professional phagocytes in echinoderms—express diverse scavenger receptors and perform opsonin-dependent engulfment, mirroring mammalian mechanisms while highlighting innovations in immune surveillance. This metazoan diversification likely arose from expansions, such as Toll-like receptors, which coordinate phagocytic responses in lacking adaptive immunity. Recent genomic analyses in the have illuminated the pre-metazoan roots of phagocytic genes, particularly in choanoflagellates—the closest unicellular relatives of animals—which actively perform bacterivory via a microvillar collar, supported by conserved actin-based machinery. Sequencing of species like Salpingoeca rosetta has identified horizontal gene transfers contributing to endocytosis-related genes, bridging unicellular phagocytosis to metazoan immune evolution and revealing how these processes facilitated the transition to multicellularity.

References

  1. [1]
    Phagocytosis: Our Current Understanding of a Universal Biological ...
    Jun 2, 2020 · Phagocytosis is a cellular process for ingesting and eliminating particles larger than 0.5 μm in diameter, including microorganisms, foreign substances, and ...
  2. [2]
    Phagocytosis: A Fundamental Process in Immunity - PMC
    Phagocytosis is an elegant but complex process for the ingestion and elimination of pathogens, but it is also important for the elimination of apoptotic cells.
  3. [3]
    Monocytes, macrophages, dendritic cells and neutrophils: an update ...
    Phagocytes form a family of immune cells that play a crucial role in tissue maintenance and help orchestrate the immune response.
  4. [4]
    In brief: The innate and adaptive immune systems - NCBI - NIH
    Aug 14, 2023 · Phagocytes: Making germs harmless. Bacteria or viruses that enter the body can be stopped right away by phagocytes, also known as scavenger ...
  5. [5]
  6. [6]
  7. [7]
    Antiinflammatory effects of apoptotic cells - JCI
    This was reflected, for example, in the normal, almost invisible removal of >1011 neutrophils per day from the adult human circulation! Uptake of apoptotic ...
  8. [8]
    Clearance of apoptotic cells by phagocytes - Nature
    Jun 15, 2007 · Efficient clearance of apoptotic cells is a prerequisite for normal tissue homeostasis as well as immune response to injury. We have discussed ...
  9. [9]
    The origins and non-canonical functions of macrophages in ...
    May 2, 2019 · It has emerged that macrophages can influence the development, homeostasis, maintenance and regeneration of many tissues and organs.
  10. [10]
    Macrophages in Tissue Repair, Regeneration, and Fibrosis
    Mar 15, 2016 · Inflammatory monocytes and tissue-resident macrophages are key regulators of tissue repair, regeneration, and fibrosis.
  11. [11]
    Histology, Alveolar Macrophages - StatPearls - NCBI Bookshelf - NIH
    Nov 10, 2024 · Besides pathogen clearance, alveolar macrophages are responsible for removing apoptotic cells, dead epithelial cells, and cellular debris ...
  12. [12]
    Functions of Macrophages in the Maintenance of Intestinal ...
    Mar 18, 2019 · Intestinal macrophages exert their functions in the maintenance of intestinal homeostasis by shaping host-microbiota symbiosis, managing gut inflammation, ...
  13. [13]
    Review Phagocytosis: Elegant Complexity - ScienceDirect.com
    An important function of phagocytes in higher organisms is to provide antigenic ligands to stimulate clonal expansion of T and B cells. In this regard DCs are ...
  14. [14]
    Macrophages and Their Role in Atherosclerosis - PubMed Central
    Macrophages play a central role in the pathogenesis of atherosclerosis. They actively participate in LDL uptake and lipid accumulation in the arterial wall ...
  15. [15]
    Macrophages as mediators of tumor immunosurveillance - PMC
    Macrophages play a major role in recognition and clearance of foreign, aged, and damaged cells. Macrophage phagocytosis is negatively regulated via the ...
  16. [16]
    Microglia in neurodegenerative diseases: mechanism and potential ...
    Sep 22, 2023 · Here we comprehensively review the biology of microglia and the roles of microglia in neurodegenerative diseases, including Alzheimer's disease, Parkinson's ...
  17. [17]
    Microglial Phagocytosis: A Disease-Associated Process Emerging ...
    We discuss the impact of established AD risk variants on microglial phagocytosis and debris processing via the endolysosomal system.
  18. [18]
    The historical milestones in the understanding of leukocyte biology ...
    May 31, 2011 · In 1867, Julius Friedrich Cohnheim (1839–1884), a German pathologist, proved that white blood cells cross blood vessels to become pus cells [33] ...
  19. [19]
  20. [20]
    Ilya Mechnikov – Nobel Lecture - NobelPrize.org
    When the anthrax bacillus is injected under the skin of sensitive animals, such as the rabbit or the guinea-pig, the microbe is found free in abundant fluid ...
  21. [21]
    The Phagocyte, Metchnikoff, and the Foundation of Immunology
    Metchnikoff described for the first time a number of crucial features of phagocytic cells, including (i) phagocyte-mediated host protection; (ii) active ...
  22. [22]
    Elie Metchnikoff, the Man and the Myth - PMC
    Feb 3, 2016 · Elie Metchnikoff, the father of innate immunity and discoverer of the significance of phagocytosis in development, homeostasis and disease.
  23. [23]
    Dynamics of phagocytosis mediated by phosphatidylserine - PMC
    Oct 25, 2022 · Phagocytosis triggered by the phospholipid phosphatidylserine (PS) is key for the removal of apoptotic cells in development, tissue homeostasis and infection.Recognition Of... · Signalling Pathways... · Figure 2. Signalling...
  24. [24]
    Generation of membrane structures during phagocytosis and ...
    This review will focus on the mechanisms by which macrophages regulate actin polymerization through initial receptor signaling and subsequent Arp2/3 activation.
  25. [25]
    Physical Constraints and Forces Involved in Phagocytosis - PMC - NIH
    Jun 12, 2020 · Phagosome formation is driven by a protrusive force (red arrows), generated by the polymerization of actin filaments, directed along the ...
  26. [26]
    Complement receptor 3 mediates both sinking phagocytosis ... - NIH
    The endocytic pathway during sinking phagocytosis is restricted in diameter and hRBCs, which are highly deformable, are squeezed into the cell interior, ...
  27. [27]
    Creatine Kinase–Mediated ATP Supply Fuels Actin-Based Events in ...
    Mar 11, 2008 · Cdc42, Rac1, and Rac2 display distinct patterns of activation during phagocytosis. Mol Biol Cell. 2004;15:3509–3519. doi: 10.1091/mbc.E03-11 ...
  28. [28]
    Regulation of phagocytosis by Rho GTPases - PMC - NIH
    All forms of phagocytosis require F-actin recruitment beneath tethered particles and F-actin re-arrangement promoting engulfment, which are controlled by Rho ...
  29. [29]
    Diversity and Versatility of Phagocytosis: Roles in Innate Immunity ...
    May 23, 2017 · In addition, phagocytes play a part in tissue remodeling and maintain overall homeostasis by disposing of apoptotic cells, a task shared by non- ...
  30. [30]
    Formation and Maturation of the Phagosome: A Key Mechanism in ...
    Unlike FcR-mediated phagocytosis to form pseudopods, CR-mediated phagocytosis is a 'sinking' phagocytosis process that internalizes foreign pathogens through ...2.2. Phagocytic Receptor... · 3. Phagosome Maturation · 3.1. Early Phagosome<|control11|><|separator|>
  31. [31]
    Patterns, Receptors, and Signals: Regulation of Phagosome ... - NIH
    During phagolysosomal fusion, the organelle acquires hydrolytic enzymes, including glycosidases, lipases, DNAses, and proteases, such as cathepsins, which ...
  32. [32]
    Control of Phagocytosis by Microbial Pathogens - PMC
    Phagocytosis can be divided into several main steps: (i) microbial recognition, (ii) phagosome formation, and (iii) phagolysosome maturation.
  33. [33]
    The Phagocyte NOX2 NADPH Oxidase in microbial killing and cell ...
    Jul 11, 2019 · Precise mechanisms of oxidant-dependent killing by phagocytes are unknown. NOX2 NADPH-generated oxidants participate in cell signaling. More ...
  34. [34]
    Oxidative killing of microbes by neutrophils - ScienceDirect.com
    Neutrophils and other phagocytic leukocytes contain a phagocyte NADPH oxidase enzyme that generates superoxide after cell activation.<|control11|><|separator|>
  35. [35]
    How Neutrophils Kill Microbes - PMC - PubMed Central - NIH
    Neutrophils provide the first line of defense of the innate immune system by phagocytosing, killing, and digesting bacteria and fungi.Missing: independent | Show results with:independent
  36. [36]
    Oxygen-Independent Bactericidal Systems: Mechanisms and ...
    During phagocytosis, granules fuse to phagolysosomes where the granule contents contribute to the destruction and dissolution of ingested bacteria.
  37. [37]
    Oxygen-dependent and oxygen-independent mechanisms of ...
    In this brief review we focus on the PMN because its antimicrobial systems have been analyzed most extensively. However, the killing mechanisms of mononuclear ...
  38. [38]
    Phagocytosis in Cellular Defense and Nutrition: A Food-Centered ...
    Intensive research has revealed in detail molecular and cellular mechanisms of phagocytosis and intracellular digestion in macrophages.
  39. [39]
  40. [40]
  41. [41]
    Neutrophil chemoattractant receptors in health and disease - Nature
    Apr 1, 2020 · The complement activation product C5a is an exceptionally powerful mediator of inflammation and has potent neutrophil chemotactic activity. The ...
  42. [42]
    How modulating LTB4 actions restore host defense in homeostasis ...
    LTB4 is a homeostatic determinant for optimal host defense in healthy individuals by driving pleiotropic actions on phagocytes that include phagocyte chemotaxis ...
  43. [43]
    How Phagocytes Acquired the Capability of Hunting and Removing ...
    Aug 20, 2021 · A study showed that in response to an fMLP gradient, some neutrophils become polarized and migrate directionally, and they also release the ...
  44. [44]
    Moving towards a paradigm: common mechanisms of chemotactic ...
    MAPK stimulation by fMLP is greater than by other chemoattractants, such as C5a, LTB4, platelet-activating factor (PAF), and IL8, although the dynamics of ...
  45. [45]
    G Protein-coupled Receptor FPR1 as a Pharmacologic Target ... - NIH
    Formylpeptide receptor1 (FPR1) is a G protein-coupled chemoattractant receptor (GPCR) originally identified in phagocytic leucocytes and mediates cell ...
  46. [46]
    Structural basis for recognition of N-formyl peptides as pathogen ...
    Sep 5, 2022 · The formyl peptide receptor 1 (FPR1) is primarily responsible for detection of short peptides bearing N-formylated methionine (fMet) that ...
  47. [47]
    Rac1 links leading edge and uropod events through Rho and ... - NIH
    We demonstrate here that Rac1 (and not Rac2) is essential for Rho and myosin activation at the trailing edge to regulate uropod function.
  48. [48]
    The Role of the Uropod in Neutrophil Polarization and Migration
    Jul 25, 2016 · During chemotactic migration, Rac activation at the lead- ing edge induces actin polymerization and pseudopod formation. (Charest and Firtel, ...Missing: phagocyte | Show results with:phagocyte
  49. [49]
    PECAM-1 supports leukocyte diapedesis by tension-dependent ...
    May 3, 2021 · Leukocyte extravasation is an essential step during the immune response and requires the destabilization of endothelial junctions.Missing: phagocyte | Show results with:phagocyte
  50. [50]
    PECAM-1: regulator of endothelial junctional integrity - PMC
    PECAM-1 is also highly expressed at endothelial cell intercellular junctions, where it functions as a mechanosensor, as a regulator of leukocyte trafficking.The Vascular Barrier · Pecam-1 Isoforms And... · Pecam-1 As A Biosensor...Missing: phagocyte | Show results with:phagocyte
  51. [51]
    Matrix metalloproteinases modulate ameboid-like migration of ...
    Aug 1, 2013 · In vitro studies suggest that leukocytes locomote in an ameboid fashion independently of pericellular proteolysis.
  52. [52]
    Extracellular Matrix Enzymes and Immune Cell Biology - Frontiers
    This review provides a current summary of the role of ECM enzymes in immune cell migration and function and discusses opportunities and limitations
  53. [53]
    Measuring neutrophil speed and directionality during chemotaxis ...
    We found consistent average velocity of (19 ± 6 μm/min) and directionality (91.1%) between the three sources. We quantified the variability in neutrophil ...
  54. [54]
    Neutrophils: Amoeboid Migration and Swarming Dynamics in Tissues
    Apr 10, 2022 · Intravital microscopy of neutrophils in inflamed organs reveals migration patterns, which appear non-directional within the interstitial tissue ...
  55. [55]
    Neutrophils self-limit swarming to contain bacterial growth in vivo
    Intravital imaging in mouse tissues has revealed that collective-like swarming behavior underlies the formation of neutrophil aggregates in many mouse ...
  56. [56]
    Multistep Navigation and the Combinatorial Control of Leukocyte ...
    We show that, over a wide range of conditions, neutrophils can migrate “down” a local chemoattractant gradient in response to a distant gradient of a different ...
  57. [57]
    How ontogeny shapes classical monocyte phenotypes - Nature
    Jan 20, 2024 · Originating primarily from progenitors in the bone marrow, monocytes are circulating mononuclear blood phagocytes, which comprise two main ...
  58. [58]
    Tissue macrophages: origin, heterogenity, biological functions ...
    Mar 7, 2025 · In the heart, macrophages play essential roles in development, repair, and remodeling. Resident cardiac macrophages expressing MHC-II ...
  59. [59]
    Bone marrow-derived monocytes give rise to self-renewing and fully ...
    Jan 27, 2016 · Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J. Exp. Med ...
  60. [60]
    Long-term culture-expanded alveolar macrophages restore their full ...
    Feb 24, 2022 · Induction of the nuclear receptor PPAR-γ by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages ...
  61. [61]
    Drugging the efferocytosis process: concepts and opportunities
    Jun 1, 2022 · During efferocytosis, MerTK blocks NF-κB signalling in macrophages to suppress inflammatory cytokine production and macrophage M1-like ...
  62. [62]
    Macrophages in cardiovascular diseases: molecular mechanisms ...
    May 31, 2024 · In terms of inflammation, macrophages facilitate chronic vascular inflammation by releasing pro-inflammatory cytokines such as IL-1, IL-6, and ...
  63. [63]
    Macrophages in immunoregulation and therapeutics - Nature
    May 22, 2023 · Macrophages are crucial in innate immunity by regulating several homeostatic and evolutionary host defense immune responses.
  64. [64]
    Trained immunity: induction of an inflammatory memory in disease
    Oct 14, 2025 · Signatures of epigenetic memory are found in monocytes of individuals post severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ...
  65. [65]
    Histology, White Blood Cell - StatPearls - NCBI Bookshelf
    Nov 14, 2022 · Neutrophils comprise 50% to 70% of circulating leukocytes and represent the body's initial line of defense. They are involved in the acute ...
  66. [66]
    Neutrophil: A Cell with Many Roles in Inflammation or Several Cell ...
    They differ in cytokine production, macrophage activation potential, expression of TLR, and expression of surface molecules. These subsets were named PMN-1 and ...Neutrophil Life Cycle · Neutrophil Exit From The... · Migrating Neutrophil...
  67. [67]
    Neutrophil Granule - an overview | ScienceDirect Topics
    Inasmuch as azurophilic granules are the first to form during granulopoiesis, they are also known as primary granules. The primary granules are the sole ...
  68. [68]
    The Role of Neutrophils in the Immune System: An Overview - NIH
    Neutrophils are the primary mediators of the rapid innate host defense against most bacterial and fungal pathogens that occurs before the complex humoral and ...
  69. [69]
    Neutrophil extracellular traps in homeostasis and disease - Nature
    Sep 20, 2024 · Collectively, NETs stimulate neutrophil effector function and bolster antimicrobial defense. Moreover, NETs possess the capacity to connect ...
  70. [70]
    Regulation of Human Neutrophil Apoptosis and Lifespan in Health ...
    Another distinguishing feature of PMNs is their short lifespan. Specifically, these cells survive for less than 24 hours in the bloodstream and are inherently ...
  71. [71]
    The role of neutrophil death in chronic inflammation and cancer
    Apr 22, 2020 · The lifespan of a neutrophil is short and limited by programmed cell death, followed by efferocytosis. When activated or exposed to insult, ...
  72. [72]
    Neutrophil extracellular traps in systemic autoimmune and ... - Nature
    Oct 18, 2022 · In this Review, we provide an overview of the roles of neutrophils in systemic autoimmune and autoinflammatory diseases and address putative therapeutic ...
  73. [73]
    Human dendritic cell subsets: an update - PMC - NIH
    Dendritic cells (DC) are a class of bone‐marrow‐derived cells arising from lympho‐myeloid haematopoiesis that form an essential interface between the innate ...
  74. [74]
    What Makes a pDC: Recent Advances in Understanding ... - Frontiers
    Plasmacytoid dendritic cells (pDCs) and two major subsets of conventional dendritic cells (cDC1 and cDC2) have been identified in mice and humans as well as ...
  75. [75]
    Molecular mechanisms of dendritic cell migration in immunity and ...
    May 25, 2020 · Immature DCs continuously sample tissues and engulf antigens via endocytic pathways such as phagocytosis or macropinocytosis, which result in DC ...Introduction · Table 1 · Dendritic Cell Migration In...
  76. [76]
    Tolerogenic dendritic cells: cytokine modulation comes of age | Blood
    Sep 1, 2006 · DC maturation is paralleled by up-regulation of major histocompatibility complex (MHC) class II and costimulatory CD80/CD86 molecules and by ...Missing: traits | Show results with:traits
  77. [77]
    Synergistic activation of dendritic cells by combined Toll-like ...
    Jul 15, 2006 · DCs are equipped with a wide variety of TLR specifically recognizing distinct microbial or viral structures. TLR ligation leads to DC activation ...Missing: traits | Show results with:traits
  78. [78]
    Deciphering tumor-infiltrating dendritic cells in the single-cell era
    Nov 27, 2023 · This review provides a comprehensive summary of the current state of knowledge regarding DC subtypes in the TME, drawing from single-cell studies conducted ...
  79. [79]
    Immuno-metabolic dendritic cell vaccine signatures associate with ...
    Nov 8, 2023 · We demonstrate that the metabolic profile of DC is tightly associated with the immunostimulatory potential of DC vaccines from cancer patients.
  80. [80]
    Non-professional phagocytosis: a general feature of normal tissue ...
    Aug 15, 2019 · Non-professional phagocytosis may participate in the limitation of these mechanisms ... We used epithelial cells, fibroblasts, endothelial cells, ...
  81. [81]
    Non-professional efferocytosis of Salmonella-infected intestinal ...
    Feb 2, 2024 · Phagocytes engulf apoptotic cells to prevent inflammation by cellular components and to recycle metabolic substrates. In the adult intestine, ...Missing: microbiome | Show results with:microbiome
  82. [82]
    Efferocytosis of vascular cells in cardiovascular disease - PMC
    In this review, we focus on macrophage efferocytosis of vascular cells, such as endothelial cells, smooth muscle cells, fibroblasts, and pericytes.
  83. [83]
    After cell death: the molecular machinery of efferocytosis - Nature
    Aug 23, 2023 · Efferocytosis is generally referred to as the phagocytosis of apoptotic cells because it involves the ingestion of extracellular substances.
  84. [84]
    Cell-Surface Calreticulin Initiates Clearance of Viable or Apoptotic ...
    Article. Cell-Surface Calreticulin Initiates Clearance of Viable or Apoptotic Cells through trans-Activation of LRP on the Phagocyte.
  85. [85]
    Efferocytosis and Its Role in Inflammatory Disorders - Frontiers
    The disposal of apoptotic cells leads to the production of IL-10 and TGF-β in macrophages, further increasing the Treg population (Kleinclauss et al., 2006).
  86. [86]
    Efferocytosis and Its Role in Inflammatory Disorders - PMC
    Efferocytosis is performed by professional phagocytes, including DCs and macrophages, and non-professional phagocytes, such as fibroblasts and epithelial cells ...
  87. [87]
    Anti-Inflammatory Neutrophils Reprogram Macrophages toward a ...
    Jan 23, 2024 · We hypothesized that N2 neutrophils can reprogram MACs toward a healing phenotype with increased efferocytosis capacity.3. Results · 3.2. Macrophages Exposed To... · 3.5. Efferocytosis Capacity...
  88. [88]
    Efferocytosis in dendritic cells: an overlooked immunoregulatory ...
    Efferocytosis, the process of engulfing and removing apoptotic cells, plays an essential role in preserving tissue health and averting undue inflammation.
  89. [89]
    Mechanisms and Consequences of Defective Efferocytosis in ... - NIH
    Efferocytosis plays a major mechanistic role in the resolution of inflammation. First, expeditious clearance of dead cells prevents their secondary necrosis.
  90. [90]
    The Role of Efferocytosis in Autoimmune Diseases - Frontiers
    Jul 19, 2018 · One of the autoimmune diseases that is strongly associated with impaired efferocytosis is SLE. This disease can cause a wide range of clinical ...
  91. [91]
    Mechanisms of efferocytosis in determining inflammation resolution ...
    Aug 26, 2022 · Efferocytosis exerts its proresolving effects by dampening proinflammatory signalling, producing proresolving mediators, modulating macrophage phenotype and ...3 Efferocytosis: A Link To... · 3.2 Targeting Spms And... · 4 Efferocytosis Triggering...
  92. [92]
    Presentation of phagocytosed antigens by MHC class I and II - PMC
    Historically, phagosomes were well recognized to provide antigens for presentation by MHC-II molecules on phagocytic cells such as macrophages and DCs before ...
  93. [93]
    Antigen Cross-Presentation by Macrophages - Frontiers
    Role of Dendritic Cells and Macrophages in CD8+ T Lymphocyte Activation. During their lifetime, DCs can exist in two discrete stages: immature and mature.
  94. [94]
    Non-equivalent antigen presenting capabilities of dendritic cells and ...
    Feb 12, 2018 · Circulating dendritic cells and macrophages have been demonstrated to be capable of antigen presentation; however, these results were acquired ...
  95. [95]
    The role of the CD28 receptor during T cell responses to antigen
    A counter-receptor for CD28 is the B7 molecule expressed on activated B cells, dendritic cells, and macrophages. B7 also binds to CTLA-4, a receptor that is ...
  96. [96]
    Costimulatory Molecules on Immunogenic Versus ... - Frontiers
    This review gives an overview of functional different human DC subsets with unique profiles of costimulatory molecules and outlines how different costimulatory ...
  97. [97]
    Interleukin-12 is produced by dendritic cells and mediates T helper 1 ...
    Interleukin-12 is produced by dendritic cells and mediates T helper 1 development as well as interferon-gamma production by T helper 1 cells.
  98. [98]
    Atomistic structure and dynamics of the human MHC-I peptide ...
    Aug 11, 2020 · We built an all-atom model of the human MHC-I PLC by combining the recent 9.9-Å resolution cryo-EM density with microsecond molecular dynamics simulations.
  99. [99]
    Self-Antigen Presentation by Dendritic Cells in Autoimmunity - PMC
    Feb 13, 2014 · The current review discusses the role of DCs in autoimmune diseases, the various factors involved in the induction and maintenance of tolerogenic DC phenotype.Missing: costimulation | Show results with:costimulation
  100. [100]
    Dendritic cells constitutively present self antigens in their immature ...
    Our results demonstrate that immature DCs constitutively present self antigens in the lymphoid organs and characterize the molecular basis of the capacity of ...
  101. [101]
    Mechanisms of immune suppression by interleukin-10 and ...
    TGF-β is an important pleiotropic cytokine with potent immunoregulatory properties and is essential for the maintenance of immunological self-tolerance in the ...
  102. [102]
    Human monocytes and macrophages regulate immune tolerance ...
    Oct 24, 2018 · Human monocytes and macrophages regulate immune tolerance via integrin αvβ8–mediated TGFβ activation. In Special Collection: Myeloid Cells 2019.
  103. [103]
    Self-antigen presentation by dendritic cells and lymphoid stroma ...
    In secondary lymphoid organs, dendritic cells (DCs) presenting self-antigen were thought to play a major role in the induction of peripheral T cell tolerance.
  104. [104]
    Intestinal CD103+ dendritic cells: master regulators of tolerance?
    CD103(+) dendritic cells (DCs) in the intestinal mucosa play a crucial role in tolerance to commensal bacteria and food antigens.
  105. [105]
    Intestinal CD103+ dendritic cells: master regulators of tolerance?
    Tolerance to food antigens relies on mucosal CD103+ dendritic cells (DCs), which promote differentiation of regulatory T (Treg) cells.
  106. [106]
    Tolerance and M2 (alternative) macrophage polarization are ... - PNAS
    Here we identify the p50 subunit of NF-κB as a key regulator of M2-driven inflammatory reactions in vitro and in vivo.Results · Endotoxin Tolerance Promotes... · Nf-κb P50 Drives The M2...
  107. [107]
    Two Faces of Macrophages: Training and Tolerance - PMC
    Nov 2, 2021 · Macrophage tolerance reduces immune responses by changes in metabolic activity and histone modifications at promoters and enhancers of ...
  108. [108]
    Efferocytosis in dendritic cells: an overlooked immunoregulatory ...
    Efferocytosis, the process of engulfing and removing apoptotic cells, plays an essential role in preserving tissue health and averting undue inflammation.
  109. [109]
    Crosstalk Between Gut Microbiota and Innate Immunity and Its ...
    Feb 20, 2020 · Gut phagocytes have pivotal roles in the maintenance of gut homeostasis, especially in the immune tolerance to symbiotic bacteria and immune ...
  110. [110]
    Understanding inborn errors of immunity - PubMed Central - NIH
    Sep 29, 2022 · IBD from underlying IEI can be categorized as: epithelial barrier defects that lead to mucosal inflammation; phagocytic defects that lead to ...
  111. [111]
    Phagocyte dysfunction and inflammatory bowel disease - PubMed
    This review consolidates the literature on inflammatory bowel disease in congenital immunodeficiencies and considers the role of phagocyte dysfunction in ...Missing: tolerance allergies
  112. [112]
    Influence of gastrointestinal commensal bacteria on the immune ...
    Jul 1, 2011 · Specifically, the microbiota can activate distinct tolerogenic dendritic cells in the gut and through this interaction can drive regulatory T- ...
  113. [113]
    from immune tolerance induction to translational challenges - PMC
    Aug 28, 2025 · Dendritic cell-derived exosomes (DEXs) have emerged as a promising tool for inducing immune tolerance and enabling precise immunomodulation in ...Missing: phagocyte | Show results with:phagocyte
  114. [114]
    Engineering M2 macrophage-derived exosomes modulate activated ...
    Through in vitro and in vivo experiments, we validate that M2CPR can induce robust and durable antigen-specific immune tolerance, offering a new paradigm for RA ...
  115. [115]
    Altered Signal Transduction in the Immune Response to Influenza ...
    ... Streptococcuspneumoniae Capsule Inhibits Complement Activity and Neutrophil Phagocytosis by Multiple Mechanisms. Infect. Immun. 2010;78:704–715. doi ...
  116. [116]
    Immunity to Infection - PMC - PubMed Central
    iv).​​ In addition, although C3b may still attach to the bacterial surface, the capsule sterically interferes with the binding of phagocyte receptors to the C3b ...
  117. [117]
    Mechanism of resistance to phagocytosis and pulmonary ...
    Mar 15, 2023 · Pseudomonas aeruginosa is known for its ability to form biofilms, which are dependent on the production of exopolysaccharides.
  118. [118]
    Pseudomonas aeruginosa Biofilms: Host Response and Clinical ...
    Sep 7, 2017 · However, despite some activity against biofilms, the function of neutrophils and macrophages is significantly attenuated by P. aeruginosa ...
  119. [119]
    Pseudomonas aeruginosa biofilm is a potent inducer of phagocyte ...
    Mar 18, 2019 · Pseudomonas aeruginosa effectively facilitate resistance to phagocyte killing by biofilm formation. However, the cross talk between biofilm components and ...
  120. [120]
    Microbial biofilm matrix restricts phagocytic cells motility in a 3D ...
    Mar 11, 2025 · Here, as proof-of-concept, we developed a 3D-microfluidics phagocyte-biofilm model to investigate how pathogenic Pseudomonas aeruginosa biofilms ...<|control11|><|separator|>
  121. [121]
    Molecular Mimicry: a Paradigm of Host-Microbe Coevolution ...
    Oct 6, 2020 · Bacterial “eukaryotic-like proteins” are a remarkable example of molecular mimicry. They are defined as proteins that strongly resemble ...
  122. [122]
    Sweet impersonators: Molecular mimicry of host glycans by bacteria
    Bacteria may exploit glycan mimics to hijack host biology to establish infection, but the glycan mimicry may also inadvertently induce abnormal immune responses ...
  123. [123]
  124. [124]
    How microorganisms avoid phagocyte attraction - Oxford Academic
    It is therefore not surprising that pathogens have evolved a large variety of strategies to evade activation of phagocytes using chemoattractants.
  125. [125]
    Yersinia pestis evades TLR4-dependent induction of IL12(p40 ... - NIH
    The data demonstrate the molecular pathway by which Y. pestis evades induction of early DC activation as measured by migration and IL12(p40) 2 production.
  126. [126]
    Virulence factors of Yersinia pestis are overcome by a strong ...
    Sep 17, 2006 · Y. pestis provides an excellent model for testing the idea that LPS modification contributes to Gram-negative virulence through evasion of TLR4 ...
  127. [127]
    Early evolutionary loss of the lipid A modifying enzyme PagP ... - PNAS
    Aug 31, 2020 · Immune evasion through membrane remodeling is a hallmark of Yersinia pestis pathogenesis. Yersinia remodels its membrane during its life cycle ...
  128. [128]
    Structural Modifications of Bacterial Lipopolysaccharide that ... - NIH
    A variety of human pathogens, including Yersinia pestis and Francisella tularensis, have lipid A moieties that are poorly recognized by the human TLR4/MD-2 ...
  129. [129]
    Evasion of the Host Immune System by Bacterial and Viral Pathogens
    Phagocytic cells have the ability to internalize microbes and kill them, as well as to recruit additional immune cells and amplify the innate response if needed ...
  130. [130]
    Immune response on biofilms – host-pathogen interactions
    This review describes the virulence factors and their interaction with the microbial communities of four well-known and highly biofilm-forming pathogens.
  131. [131]
    Control of Phagocytosis by Microbial Pathogens - Frontiers
    Protein A (SpA) (131) and staphylococcal binder of IgG (Sbi) protein specifically bind to the Fc region of IgG (132–134), blocking Fc receptor (FcR) engagement ...
  132. [132]
    M Protein of the Group A Streptococcus Binds to the Seventh Short ...
    Streptococcus pyogenes evades complement by binding the complement-regulatory protein factor H (fH) via the central conserved C-repeat region of M protein.
  133. [133]
    The Fibrinogen-binding M1 Protein Reduces Pharyngeal Cell ...
    The surface-anchored GAS M1 protein is a classical virulence factor that promotes phagocyte resistance and exaggerated inflammation by binding host fibrinogen ( ...
  134. [134]
    SopE and SopE2 from Salmonella typhimurium Activate Different ...
    Thus, selective activation of specific RhoGTPases leads to specific cellular responses. Invasion of the Gram-negative bacterial enteropathogen Salmonella ...<|separator|>
  135. [135]
    Hijacking Rho GTPases by protein toxins and apoptosis - Nature
    Apr 16, 2003 · Mimicry of Rho regulatory proteins GEF and GAP by Salmonella type-III-secreted toxins (SopE and SptP) results in prophagocytic activity that ...
  136. [136]
    Mycobacterium tuberculosis protein tyrosine phosphatase (PtpA ...
    Mycobacterium tuberculosis (Mtb) pathogenicity depends on its ability to inhibit phagosome acidification and maturation processes after engulfment by ...
  137. [137]
    Inhibition of Phagocytosis in HIV-1-infected Macrophages Relies on ...
    May 27, 2010 · Here we show that phagocytosis by various receptors was inhibited in primary human macrophages infected with wild-type HIV-1 but not with a nef-deleted virus.
  138. [138]
    Protein F1 and Streptococcus pyogenes Resistance to Phagocytosis
    Both intergenic recombination and horizontal gene transfer have played a role in generating variability in the prtF1 or sfbI gene and the emm gene (25, 29).
  139. [139]
    The Legionnaires' disease bacterium (Legionella pneumophila ...
    The Legionnaires' disease bacterium (Legionella pneumophila) inhibits phagosome-lysosome fusion in human monocytes. Copyright and License information.Missing: Sid effectors
  140. [140]
    Structure of the Legionella Virulence Factor, SidC Reveals a Unique ...
    L. pneumophila delivers nearly 300 effector proteins into host cells for the establishment of a replication-permissive compartment known as the Legionella- ...<|separator|>
  141. [141]
    The Dot/Icm type IV secretion system of Legionella pneumophila is ...
    We have previously shown that Legionella pneumophila induces caspase 3-dependent apoptosis in mammalian cells during early stages of infection.
  142. [142]
    The distinctive roles played by the superoxide dismutases ... - Nature
    Mar 12, 2022 · Salmonella enterica serovar Typhimurium periplasmic superoxide dismutases SodCI and SodCII are required for protection against the phagocyte ...
  143. [143]
    A Comprehensive Assessment of the Genetic Determinants ... - Nature
    Dec 6, 2017 · An essential mechanism uses by the phagocytes to kill and eradicate Salmonella is production of reactive oxygen species (ROS). Hydrogen peroxide ...
  144. [144]
    Listeriolysin O: a phagosome-specific lysin - PubMed - NIH
    Listeriolysin O (LLO) is a pore-forming toxin that ruptures the phagosomal membrane, allowing Listeria to access the host cytosol.
  145. [145]
    Listeriolysin O: A phagosome-specific cytolysin revisited - PubMed
    Listeriolysin O (LLO) is essential for Listeria to escape host cell vacuoles, allowing replication in the cytosol. It is a pore-forming toxin.
  146. [146]
  147. [147]
    Infection by Mycobacterium tuberculosis promotes human alveolar ...
    The induction of apoptosis in alveolar macrophages by M. tuberculosis may play a role in the macrophage-pathogen interaction of tuberculosis in vivo.Missing: examples | Show results with:examples
  148. [148]
    Leishmania amazonensis sabotages host cell SUMOylation for ...
    Aug 13, 2022 · These virulence mechanisms inhibit host cell defense responses and generate a specialized replicative niche, the parasitophorous vacuole.
  149. [149]
    Intracellular trafficking and the parasitophorous vacuole of ... - PubMed
    These findings significantly extend our understanding of the mechanisms employed by Leishmania to ensure its survival in the macrophage.
  150. [150]
    PDCD6 regulates lactate metabolism to modulate LC3-associated ...
    Nov 23, 2024 · Here, we found that intracellular L. monocytogenes were mostly retained within single-membrane phagosomes, and fewer bacteria escaped in Pdcd6 ...
  151. [151]
    Pathogenesis, immunology, and diagnosis of latent Mycobacterium ...
    Phagocytosis of tubercle bacilli by antigen-presenting cells in human lung alveoli initiates a complex infection process by Mycobacterium tuberculosis and a ...
  152. [152]
    Latent tuberculosis infection: myths, models, and ... - PubMed
    The aim of this review is to present the current state of knowledge on human latent tuberculosis infection (LTBI) based on clinical studies and observations.
  153. [153]
    From Flies to Men: ROS and the NADPH Oxidase in Phagocytes
    These pathways require tight regulation, as excessive ROS produced by phagocytes may cause oxidative stress and damage in the tissues and contribute to e.g., ...
  154. [154]
    Regulating ROS | Nature Reviews Immunology
    May 6, 2014 · Through NRROS, the production of ROS by phagocytes can be fine-tuned to control invading pathogens while limiting collateral tissue damage.
  155. [155]
    Role of elastases in the pathogenesis of chronic obstructive ...
    Neutrophil elastase, metalloproteinases, and their inhibitors play an important role in the development of chronic obstructive pulmonary disease (COPD).
  156. [156]
    Adverse effects of neutrophils on the lung - PubMed
    Accordingly, defects in regulatory mechanisms allow neutrophils to damage the lungs. Emphysema serves as a prime example of this link between host defense and ...
  157. [157]
    Cytokine Storm | New England Journal of Medicine
    Dec 2, 2020 · Cytokine storm and cytokine release syndrome are life-threatening systemic inflammatory syndromes involving elevated levels of circulating cytokines and immune ...
  158. [158]
    Deep insight into cytokine storm: from pathogenesis to treatment
    Apr 16, 2025 · Macrophages infected in conditions such as sepsis and HLH can trigger cytokine storm, during which the synergistic stimulation by ...
  159. [159]
    Cells of the synovium in rheumatoid arthritis. Macrophages
    Dec 21, 2007 · Macrophages (Mφ) are of central importance in rheumatoid arthritis (RA) due to their prominent numbers in the inflamed synovial membrane and at ...<|separator|>
  160. [160]
    Synovial Mononuclear Phagocytes in Rheumatoid Arthritis and ...
    Results demonstrate that synovial tissue from RA patients (as compared to that from OA patients) weighed more, contained more cells, more macrophages, and more ...
  161. [161]
    Reperfusion injury and reactive oxygen species - PubMed Central
    This review provides a synopsis of the evidence implicating ROS in reperfusion injury, the clinical implications of this phenomenon, and summarizes current ...
  162. [162]
    Ischemia-reperfusion injury: molecular mechanisms and therapeutic ...
    Jan 8, 2024 · Ischemia-reperfusion (I/R) injury paradoxically occurs during reperfusion following ischemia, exacerbating the initial tissue damage.
  163. [163]
    Chronic Granulomatous Disease - Medscape Reference
    Jul 29, 2019 · Chronic granulomatous disease (CGD) is a primary immunodeficiency caused by defects in any of the five subunits of the NADPH oxidase complex ...Practice Essentials · Background · Pathophysiology · Etiology
  164. [164]
    Clinical presentation, diagnosis, and treatment of chronic ... - Frontiers
    Chronic granulomatous disease (CGD) is caused by an impaired respiratory burst reaction in phagocytes. CGD is an X-linked (XL) (caused by pathogenic variants ...Abstract · Molecular genetics of chronic... · Conservative vs. curative... · Summary
  165. [165]
    Polarization of Rheumatoid Macrophages by TNF Targeting ...
    Anti-TNF agents modulated the polarization of inflammatory macrophages by decreasing inflammatory surface markers (CD40, CD80) and favoring alternative markers.
  166. [166]
    Macrophages in rheumatoid arthritis
    Apr 12, 2000 · Macrophages appear to play a pivotal role in RA because they are numerous in the inflamed synovial membrane and at the cartilage-pannus junction.
  167. [167]
    Persistent complement dysregulation with signs of ... - Science
    Jan 19, 2024 · Long Covid patients exhibited increased complement activation during acute disease, which also persisted at 6-month follow-up. The complement ...
  168. [168]
    The origin of phagocytosis in Earth history | Interface Focus - Journals
    Jun 12, 2020 · Phagocytosis, or 'cell eating', is a eukaryote-specific process where particulate matter is engulfed via invaginations of the plasma membrane.
  169. [169]
    The origins of phagocytosis and eukaryogenesis - Biology Direct
    Feb 26, 2009 · The low sequence conservation is a consequence of a dramatic acceleration of evolution of actin that was triggered by the major functional ...
  170. [170]
    Dynamic Evolution of Toll-Like Receptor Multigene Families in ...
    There are four primary classes of adult sea urchin coelomocytes: phagocytes, vibratile, colorless spherule cells, and red spherule cells (Smith et al., 2011).
  171. [171]
    Prey Capture and Phagocytosis in the Choanoflagellate ...
    May 7, 2014 · In thecate cells and rosette colonies, phagocytosis initially involves fusion of multiple microvilli, followed by remodeling of the collar ...
  172. [172]
    Detection of horizontal gene transfer in the genome of the ... - Nature
    Mar 16, 2021 · We identified at least 175 candidate HGTs in the genome of the colonial choanoflagellate Salpingoeca rosetta using sequence-based tests.