Alveolar macrophage

Last updated
Micrograph showing hemosiderin-laden alveolar macrophages, as seen in a pulmonary haemorrhage. H&E stain. Pulmonary haemorrhage - very high mag.jpg
Micrograph showing hemosiderin-laden alveolar macrophages, as seen in a pulmonary haemorrhage. H&E stain.

An alveolar macrophage, pulmonary macrophage, (or dust cell) is a type of macrophage, a professional phagocyte, found in the airways and at the level of the alveoli in the lungs, but separated from their walls. [1]

Contents

Activity of the alveolar macrophage is relatively high, because they are located at one of the major boundaries between the body and the outside world. They are responsible for removing particles such as dust or microorganisms from the respiratory surfaces.

Alveolar macrophages are frequently seen to contain granules of exogenous material such as particulate carbon that they have picked up from respiratory surfaces. Such black granules may be especially common in smoker's lungs or long-term city dwellers.

The alveolar macrophage is the third cell type in the alveolus; the others are the type I and type II pneumocytes.

Comparison of pigmented pulmonary macrophages

DiseaseMacrophage nameMacrophage pigment appearance (HE stain)Usual macrophage locationAssociated medical historyImageImage comment
Anthracosis Black-brown granulesInterstitium (perivascular)
  • Age and urban dwelling. [2]
  • Coal workers [3]
Histopathology of anthracotic macrophage in lung, annotated.jpg Black arrow shows interstitial anthracotic pigment. Nearby macrophages (white arrow) can be presumed to contain anthracotic pigment.
Respiratory bronchiolitis "Smoker’s macrophages"Yellow to light brown and finely granular [4] Airways (especially respiratory bronchioles)Tobacco smoking Histopathology of a smoker's macrophage.jpg Smoker's macrophage in center
Chronic pulmonary congestion Siderophages Brown-golden and refractile. [5] Alveoli [6]
  • Heart failure
  • Pulmonary hemorrhage
Histopathology of siderophage in chronic pulmonary congestion.jpg Siderophage (black arrow), and interstitium with edema, hemosiderin deposition (black arrow) and collagenous thickening, indicating heart failure.
Histopathology of smoker's macrophages with anthracotic stippling, indicating the presence of both respiratory bronchiolitis and anthracosis. Histopathology of smoker's macrophages with anthracotic stippling.jpg
Histopathology of smoker's macrophages with anthracotic stippling, indicating the presence of both respiratory bronchiolitis and anthracosis.

Function

Micrograph of carbon-laden macrophages in the lung, H&E stain Carbon laden macrophages in lung, H&E 100X.jpg
Micrograph of carbon-laden macrophages in the lung, H&E stain
Micrograph of an alveolar macrophage in the lung tissue showing the nucleus and other organelles including the Golgi body and mitochondria. Macrophage in the alveolus Lung - TEM.jpg
Micrograph of an alveolar macrophage in the lung tissue showing the nucleus and other organelles including the Golgi body and mitochondria.

Alveolar macrophages are phagocytes that play a critical role in homeostasis, host defense, and tissue remodeling. [7] Their population density is decisive for these many processes. They are highly adaptive and can release many secretions, to interact with other cells and molecules using several surface receptors. Alveolar macrophages are also involved in the phagocytosis of apoptotic and necrotic cells. [8] They need to be selective of the material that is phagocytized to safeguard the normal cells and structures. [8] To combat infection, the phagocytes facilitate many pattern recognition receptors (PRRs) to help recognize pathogen-associated molecular patterns (PAMPs) on the surface of pathogenic microorganisms. [9] PAMPs all have the common features of being unique to a group of pathogens but invariant in their basic structure; and are essential for pathogenicity (ability of an organism to produce an infectious disease in another organism). [9] Proteins involved in microbial pattern recognition include mannose receptor, complement receptors, DC-SIGN, Toll-like receptors(TLRs), the scavenger receptor, CD14, and Mac-1. [9] [10] PRRs can be divided into three classes:

  1. signaling PRRs that activate gene transcriptional mechanisms that lead to cellular activation,
  2. endocytic PRRs that function in pathogen binding and phagocytosis, and
  3. secreted PRRs that usually function as opsonins or activators of complement.

The recognition and clearance of invading microorganisms occurs through both opsonin-dependent and opsonin–independent pathways. The molecular mechanisms facilitating opsonin-dependent phagocytosis are different for specific opsonin/receptor pairs. For example, phagocytosis of IgG-opsonized pathogens occurs through the Fcγ receptors (FcγR), and involves phagocyte extensions around the microbe, resulting in the production of pro-inflammatory mediators. Conversely, complement receptor-mediated pathogen ingestion occurs without observable membrane extensions (particles just sink into the cell) and does not generally results in an inflammatory mediator response.

Following internalization, the microbe is enclosed in a vesicular phagosome which then undergoes fusion with primary or secondary lysosomes, forming a phagolysosome. [9] There are various mechanisms that lead to intracellular killing; there are oxidative processes, and others independent of the oxidative metabolism. The former involves the activation of membrane enzyme systems that lead to a stimulation of oxygen uptake (known as the respiratory burst), and its reduction to reactive oxygen intermediates (ROIs), molecular species that are highly toxic for microorganisms. [9] The enzyme responsible for the elicitation of the respiratory burst is known as nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, which is composed of five subunits. [9] One component is a membrane cytochrome made up of two protein subunits, gp91phox and p22phox; the remaining three components are cytosolic-derived proteins: p40phox, p47phox, and p67phox. [9] NADPH oxidase exists in the cytosol of the AM when in a quiescent state; but upon activation, two of its cytosolic components, p47phox and p67phox, have their tyrosine and serine residues phosphorylated, which are then able to mediate translocation of NADPHox to the cytochrome component, gp91phox/p22phox, on the plasma membrane via cytoskeletal elements. [11]

Compared to other phagocytes, the respiratory burst in AM is of a greater magnitude. [9] Oxygen-independent microbicidal mechanisms are based on the production of acid, on the secretion of lysozymes, on iron-binding proteins, and on the synthesis of toxic cationic polypeptides. [9] Macrophages possess a repertoire of antimicrobial molecules packaged within their granules and lysosomes. [9] These organelles contain a myriad of degradative enzymes and antimicrobial peptides that are released into the phagolysosome, such as proteases, nucleases, phosphatases, esterases, lipases, and highly basic peptides. [9] Moreover, macrophages possess a number of nutrient deprivation mechanisms that are used to starve phagocytosed pathogens of essential micronutrients. [9] Certain microorganisms have evolved countermeasures which enable them to evade being destroyed by phagocytes. Although lysosomal-mediated degradation is an efficient means by which to neutralize an infection and prevent colonization, several pathogens parasitize macrophages, exploiting them as a host cell for growth, maintenance and replication. [9] Parasites like Toxoplasma gondii and mycobacteria are able to prevent fusion of phagosomes with lysosomes, thus escaping the harmful action of lysosomal hydrolases. Others avoid lysosomes by leaving the phagocytic vacuole, to reach the cytosolic matrix where their development is unhindered. In these instances, macrophages may be triggered to actively destroy phagocytosed microorganisms by producing a number of highly toxic molecules and inducing deprivational mechanism to starve it. [9] Finally, some microbes have enzymes to detoxify oxygen metabolites formed during the respiratory burst. [9]

When insufficient to ward off the threat, alveolar macrophages can release proinflammatory cytokines and chemokines to call forth a highly developed network of defensive phagocytic cells responsible for the adaptive immune response.

The lungs are especially sensitive and prone to damage, thus to avoid collateral damage to type 1 and type II pneumocytes, alveolar macrophages are kept in a quiescent state, producing little inflammatory cytokines and displaying little phagocytic activity, as evidenced by downregulated expression of the phagocytic receptor Macrophage 1 antigen (Mac-1). [7] [12] AMs actively suppress the induction of two of the immunity systems of the body: the adaptive immunity and humoral immunity. The adaptive immunity is suppressed through AM's effects on interstitial dendritic cells, B-cells and T-cells, as these cells are less selective of what they destroy, and often cause unnecessary damage to normal cells. To prevent uncontrolled inflammation in the lower respiratory tract, alveolar macrophages secrete nitric oxide, prostaglandins, interleukin-4 and -10(IL-4, IL-10), and transforming growth factor-β (TGF-β). [12] [13] [14] [15]

Role of signaling molecules

Nitric oxide

Nitric oxide (NO) is a major source of immunomodulation in rodents, and is produced by enzyme nitric oxide synthetase type 2 (NOS2) in the alveolar macrophage. [14] NO inhibits tyrosine phosphorylation of the kinases involved in production of the interleukin-2 (IL-2) receptor, the expression of which is fundamental for T cell proliferation. [13] In humans, however, NOS2 activity has been difficult to verify. [14]

There are two explanations for the lack of responsiveness in the promoter of human inducible nitric oxide synthetase (iNOS) to NO activation by lipopolysaccharides (LPS) + interferon gamma (IFNγ). [14] The first is that there are various inactivating nucleotide variations in the human counterpart of the enhancer element that regulates LPS/IFNγ induced expression of the mouse NOS2 gene. The second is because of the absence of a nuclear factor in human macrophages that is required for optimum expression of gene NOS2 (LPS-inducible nuclear factor-kappa B/Rel complex). [14] It is assumed that the difficulty in verifying NOS2 is due to a much more tightly controlled expression in human AMs as compared to that in the rodent AMs. [14] NOS2 is part of an autoregulatory feedback loop, wherein an allergen or provoker stimulates inflammatory cytokine production, which in turn stimulates NO production, and NO down-regulates cytokine production. [14] In rats, NO inhibits the granulocyte-macrophage colony-stimulating factor (GM-CSF)-mediated maturation of dendritic cells, and in humans it inhibits the TNF-alpha-mediated maturation of human dendritic cells, through cyclic GMP-dependent mechanisms. [14] NO prolongs the ability of human dendritic cells to internalize antigens at sites of inflammation, therefore modulating the beginning steps leading to antigen-specific immune responses. [14]

NO production has been implicated as relevant to the pathology of asthma. People with asthma show an increased expression of iNOS in airway epithelial cells and an increased level of nitric oxide in exhaled air. [14]

Prostaglandin endoperoxide 2 (PGE2)

Many other immunomodulating factors have been isolated, the most important of which are prostaglandins and cytokines. PGE2 was the first immunomodulator to be derived from macrophages and described. [14] PGE2 functions in amplifying peripheral blood lymphocyte IL-10 transcription and protein production; as well as in deactivating macrophages and T-cells. [14] PGE2 is an immunomodulatory eicosanoid derived from the cell membrane component, arachidonic acid, and is processed in the arachidonic acid cascade: the successive oxygenation and isomerization of arachidonic acid by cyclooxygenase and PGE2 synthase enzymes. [16] The regulation of target cells by PGE2 occurs via signaling through four cell membrane-associated G-protein-coupled E-prostanoid (EP) receptors, named EP1, EP2, EP3, and EP4. [16] PGE2 inhibits bacterial killing and ROI production by AM by impairing Fcγ-mediated phagocytosis through its ability to stimulate the production of intracellular cyclic adenosine monophosphate (cAMP) effectors via EP2 and EP4 receptors signaling. [11] [16] EP2 and EP4 receptors signal primarily through stimulatory G protein (Gs), increasing adenylyl cyclase (AC) activity and subsequent cAMP formation. [11] cAMP is a second messenger that influences multiple cellular functions via the activation of two downstream effector molecules, protein kinase A (PKA) and the exchange proteins directly activated by cAMP (Epac-1 and -2). [11] Epac-1 and PKA are both important factors involved in the inhibition of AM bacterial killing. [11] The effects of PKA results from its ability to phosphorylate serine and threonine residues on many cellular proteins, especially transcription factor cAMP response element binding protein (CREB). cAMP/PKA/CREB axis mediates the inhibition of TNF-alpha release. [11] The killing of phagocytosed bacteria by AMs is dependent upon several distinct microbicidal mechanisms, like the reduced NADPH oxidase-mediated release of ROI. [9] [11] ROI generation by NADPH oxidase is an important bactericidal mechanism after FcR-mediated phagocytosis. [11] PGE2 activates both Gs-coupled EP2 and EP4 receptors by ligation, stimulating cAMP production and subsequent activation of downstream cAMP effectors, PKA and Epac-1; both which in turn impair the phosphorylation and phagosomal membrane translocation of NADPH oxidase component, p47phox, thereby inhibiting the respiratory burst. [11]

Interleukin-4 and -10

IL-4 is a pleiotropic cytokine that plays a key role in the development of T helper type 2(Th2) cells. IL-4 is important for the differentiation of naïve CD4-T cells into mature Th2 type cells; as well as for Immunoglobulin (Ig) class switching to IgE and IgG4 during the development of immune responses. [17] [18] Ig is a class of antibody found only in mammals that plays an important role in allergy response and defense against many kinds of pathogens by protecting the body against them by complement activation, opsonization for phagocytosis, and neutralization of their toxins. [18]

IL-4 and IL-10 have both been shown to reduce the production of metalloproteinases (endopeptidases which break down collagen and other extracellular proteins) by human AMs. [14] [15] IL-4 has dual effects upon macrophage biological function, which may be either stimulatory or inhibitory. [15] It enhances MHC class II antigen (extracellular protein complex that interacts exclusively with CD4-T cells as part of the exogenous pathway) and Mac-1(surface receptor as part of innate complement system) expression, thus promoting phagocytosis. [15] IL-4 has also been shown to inhibit the production of PGE2 by reducing the expression of the enzyme, prostaglandin H synthase -2 (PGHS-2), which is critical in the production of PGE2. [14] However, IL-4 inhibits production of TNF-alpha, IL-1 and -6, which are all important cytokines in the proinflammatory response). [15]

IL-10 inhibits the secretion of pro-inflammatory cytokines TNF-alpha and INF-gamma, thus suppressing the proliferation of T-cells, NK cells, and AM. [14] IL-10 shares similar immunomodulating mechanisms to TGF-β. [14] It is thought that both cytokines reduce the rate of apoptosis in human alveolar macrophages, thus indirectly enhancing alveolar macrophage-mediated inhibition of T-cell proliferation. [14] There is a significant increase in the basal rate of apoptosis upon activation by bacterial products. Apoptosis is particularly regulated by the presence of cytokines: IFNγ increases the rate of apoptosis, whereas IL-10 and TGF-β decrease it. [14] However, IL-10 has counterproductive effects on the immune system, and has been shown to actually promote infection by foreign pathogens. The role of IL-10 in bacterial and parasitic infection has been discovered as a strategy to evade host immune systems. [19] There are bacteria which parasitize AMs by invading through their membranes, and thrive by growing and replicating inside of them, exploiting AMs as host cells. Normally, this infection can be eliminated by T-cells, which activate enzymes in alveolar macrophages that destroy the bacteria; but these bacteria have been shown to alter the cytokine signaling network to their advantage. As an inhibitory cytokine, IL-10 facilitates the infection of human alveolar macrophages and monocytes by completely reversing the protective effect of IFNγ against intracellular Legionella pneumophila replication. [19] Yersinia enterocolitica has also been shown to releases virulence antigen LcrV, which induces IL-10 through Toll-like receptor-2 and CD14 (an accessory surface protein of TLR4-mediated LPS-signaling), resulting in the suppression of IFNγ and TNF-alpha suppression. [19]

Transforming growth factor β (TGF-β)

In normal conditions, alveolar macrophages adhere closely to alveolar epithelial cells, thus inducing the expression of the αvβ6 integrin. Integrins are dimeric cell-surface receptors composed of alpha and beta subunits, which activates TGF-β.< [20] [21] TGF-β is a multifunctional cytokine that modulates a variety of biological processes such as cell growth, apoptosis, extracellular matrix synthesis, inflammation, and immune responses. [22] TGF-β tightly regulates anti-inflammatory activity by suppressing pro-inflammatory cytokine production, thereby inhibiting T-lymphocyte function. [23] Integrins avβ6 and avβ8 sequester latent TGF-β to the cell surface, where activation can be tightly coupled to cellular responses to environmental stress in the maintenance of homeostasis; integrins also localize activated TGFβ in the vicinity of the macrophages. [24] Normally mature TGFβ is secreted as a latent complex with its N-terminal fragment, latency-associated peptide (LAP), which inhibits its activity. [22] The latent complex is covalently linked to the extracellular matrix by binding to latent TGF-β-binding proteins. [20] TGF-β is activated by diverse mechanisms in the lung, ultimately involving either proteolysis or conformational alteration of the LAP. [24] αvβ6 integrin is able to mediate activation of TGF-β by binding to TGF-β1 LAP, which serves as a ligand binding site for the integrin, and is an essential component of the TGF-β activation apparatus. [22] [25] Once activated, TGFβ leads to the suppression of macrophage functionality (cytokine production and phagocytosis). [22] Binding of activated TGF-β to its receptors expressed on alveolar macrophages induces a downstream signaling cascade, including phosphorylation of receptor-regulated Small Mothers Against Decapentaplegic (R-SMAD)homologs 2 and 3. [7] [22] [23] Phosphorylated SMAD-2 and -3 then form heteromeric complexes with common-mediator SMAD 4 (co-SMAD-4). Once assembled, the complexes translocates into the nucleus via the nuclear pore with the assistance of importins alpha/beta. Once in the nucleus, these complexes accumulate and eventually act as a transcription factors, regulating the expression of TGF-β target genes. [23] Thus TGF-β signaling involves a direct pathway from the receptors on the surface of a cell to the nucleus.

Activation

Toll-like receptors (TLRs) are signaling PRRs, capable of recognizing various bacterial proteins. [10] Although bacteria have evolved means of evading host defense mechanisms, they express PAMPs, such as lipoglycans and lipoproteins that are recognized by cells of the innate immune system through the TLRs. [10] Upon binding of PAMPs to TLRs, the TLR triggers inflammatory and defensive responses in the host cell, inducing actin polymerization in alveolar macrophages (a crucial component in endocytosis and motility). [22] Actin polymerization in alveolar macrophages causes the suppression of integrin expression, which in turn causes the deactivation of TGF-β and the downregulation of the basal phosphorylation level of SMAD 2/3; subsequently leading to the activation and detachment of alveolar macrophages from the alveolar epithelial cells [22] [15]. Upon activation, macrophages become primed for phagocytosis and begin to secrete proinflammatory cytokines (TNF-α and IL-6). [22]

The priming of macrophages involves the enhancement of respiratory burst activity by IFN-γ and TNF-α. [9] IFNγ induces both an increased affinity of the NADPH oxidase for NADPH in macrophages, as well as an increased rate of gene transcription and message expression for gp91phox protein. [9] TNF-α acts as an autocrine stimulus by increasing the expression of both p47phox and p67phox transcripts. The ROIs produced during the respiration burst response, in turn, enhance production of TNF-α by macrophages. [9]

Deactivation

Gas exchange must be restored as quickly as possible to avoid collateral damage, so activated lymphocytes secrete IFNγ to stimulate the production of matrix metalloproteinase MMP-9 by macrophages. [22] AMs have been reported to produce MMP-9 partly via PGE2-dependent PKA signaling pathways, which are the pathways involved in the inhibition of phagocytosis. [26] MMP-9 activates latent TGF-β, reinducing expression of αvβ6 integrins on alveolar epithelial cells, thereby returning the alveolar macrophage to a resting state. [7] [22] [26] Activation of TGF-β is also advantageous because its production stimulates collagen synthesis in interstitial fibroblasts, which is necessary for restoring alveolar wall architecture. [22]

See also

Related Research Articles

<span class="mw-page-title-main">Cytokine</span> Broad and loose category of small proteins important in cell signaling

Cytokines are a broad and loose category of small proteins important in cell signaling. Due to their size, cytokines cannot cross the lipid bilayer of cells to enter the cytoplasm and therefore typically exert their functions by interacting with specific cytokine receptors on the target cell surface. Cytokines have been shown to be involved in autocrine, paracrine and endocrine signaling as immunomodulating agents.

<span class="mw-page-title-main">Macrophage</span> Type of white blood cell

Macrophages are a type of white blood cell of the innate immune system that engulf and digest pathogens, such as cancer cells, microbes, cellular debris, and foreign substances, which do not have proteins that are specific to healthy body cells on their surface. This process is called phagocytosis, which acts to defend the host against infection and injury.

<span class="mw-page-title-main">Phagocytosis</span> Process by which a cell uses its plasma membrane to engulf a large particle

Phagocytosis is the process by which a cell uses its plasma membrane to engulf a large particle, giving rise to an internal compartment called the phagosome. It is one type of endocytosis. A cell that performs phagocytosis is called a phagocyte.

<span class="mw-page-title-main">Tumor necrosis factor</span> Protein

Tumor necrosis factor is an adipokine and a cytokine. TNF is a member of the TNF superfamily, which consists of various transmembrane proteins with a homologous TNF domain.

<span class="mw-page-title-main">Phagocyte</span> Cells that ingest harmful matter within the body

Phagocytes are cells that protect the body by ingesting harmful foreign particles, bacteria, and dead or dying cells. Their name comes from the Greek phagein, "to eat" or "devour", and "-cyte", the suffix in biology denoting "cell", from the Greek kutos, "hollow vessel". They are essential for fighting infections and for subsequent immunity. Phagocytes are important throughout the animal kingdom and are highly developed within vertebrates. One litre of human blood contains about six billion phagocytes. They were discovered in 1882 by Ilya Ilyich Mechnikov while he was studying starfish larvae. Mechnikov was awarded the 1908 Nobel Prize in Physiology or Medicine for his discovery. Phagocytes occur in many species; some amoebae behave like macrophage phagocytes, which suggests that phagocytes appeared early in the evolution of life.

<span class="mw-page-title-main">Cell-mediated immunity</span> Immune response that does not involve antibodies

Cell-mediated immunity or cellular immunity is an immune response that does not involve antibodies. Rather, cell-mediated immunity is the activation of phagocytes, antigen-specific cytotoxic T-lymphocytes, and the release of various cytokines in response to an antigen.

<span class="mw-page-title-main">Interleukin 12</span> Interleukin

Interleukin 12 (IL-12) is an interleukin that is naturally produced by dendritic cells, macrophages, neutrophils, and human B-lymphoblastoid cells (NC-37) in response to antigenic stimulation. IL-12 belongs to the family of interleukin-12. IL-12 family is unique in comprising the only heterodimeric cytokines, which includes IL-12, IL-23, IL-27 and IL-35. Despite sharing many structural features and molecular partners, they mediate surprisingly diverse functional effects.

<span class="mw-page-title-main">Phagosome</span>

In cell biology, a phagosome is a vesicle formed around a particle engulfed by a phagocyte via phagocytosis. Professional phagocytes include macrophages, neutrophils, and dendritic cells (DCs).

<span class="mw-page-title-main">Transforming growth factor beta</span> Cytokine

Transforming growth factor beta (TGF-β) is a multifunctional cytokine belonging to the transforming growth factor superfamily that includes three different mammalian isoforms and many other signaling proteins. TGFB proteins are produced by all white blood cell lineages.

<span class="mw-page-title-main">Innate immune system</span> One of the two main immunity strategies

The innate, or nonspecific, immune system is one of the two main immunity strategies in vertebrates. The innate immune system is an alternate defense strategy and is the dominant immune system response found in plants, fungi, insects, and primitive multicellular organisms.

<span class="mw-page-title-main">TGF beta 1</span> Protein-coding gene in the species Homo sapiens

Transforming growth factor beta 1 or TGF-β1 is a polypeptide member of the transforming growth factor beta superfamily of cytokines. It is a secreted protein that performs many cellular functions, including the control of cell growth, cell proliferation, cell differentiation, and apoptosis. In humans, TGF-β1 is encoded by the TGFB1 gene.

<span class="mw-page-title-main">Interleukin 19</span> Protein-coding gene in the species Homo sapiens

Interleukin 19 (IL-19) is an immunosuppressive protein that belongs to the IL-10 cytokine subfamily.

Understanding of the antitumor immunity role of CD4+ T cells has grown substantially since the late 1990s. CD4+ T cells (mature T-helper cells) play an important role in modulating immune responses to pathogens and tumor cells, and are important in orchestrating overall immune responses.

Macrophage-1 antigen is a complement receptor ("CR3") consisting of CD11b and CD18.

<span class="mw-page-title-main">CLEC7A</span> Protein-coding gene in humans

C-type lectin domain family 7 member A or Dectin-1 is a protein that in humans is encoded by the CLEC7A gene. CLEC7A is a member of the C-type lectin/C-type lectin-like domain (CTL/CTLD) superfamily. The encoded glycoprotein is a small type II membrane receptor with an extracellular C-type lectin-like domain fold and a cytoplasmic domain with a partial immunoreceptor tyrosine-based activation motif. It functions as a pattern-recognition receptor for a variety of β-1,3-linked and β-1,6-linked glucans from fungi and plants, and in this way plays a role in innate immune response. Expression is found on myeloid dendritic cells, monocytes, macrophages and B cells. Alternate transcriptional splice variants, encoding different isoforms, have been characterized. This gene is closely linked to other CTL/CTLD superfamily members on chromosome 12p13 in the natural killer gene complex region.

<span class="mw-page-title-main">CD200</span> Protein-coding gene in the species Homo sapiens

OX-2 membrane glycoprotein, also named CD200 is a human protein encoded by the CD200 gene. CD200 gene is in human located on chromosome 3 in proximity to genes encoding other B7 proteins CD80/CD86. In mice CD200 gene is on chromosome 16.

An inflammatory cytokine or proinflammatory cytokine is a type of signaling molecule that is secreted from immune cells like helper T cells (Th) and macrophages, and certain other cell types that promote inflammation. They include interleukin-1 (IL-1), IL-6, IL-12, and IL-18, tumor necrosis factor alpha (TNF-α), interferon gamma (IFNγ), and granulocyte-macrophage colony stimulating factor (GM-CSF) and play an important role in mediating the innate immune response. Inflammatory cytokines are predominantly produced by and involved in the upregulation of inflammatory reactions.

Adenylate cyclase toxin (CyaA) is released from bacterium Bordetella pertussis by the T1SS and released in the host’s respiratory tract in order to suppress its early innate and subsequent adaptive immune defense.

Regulatory macrophages (Mregs) represent a subset of anti-inflammatory macrophages. In general, macrophages are a very dynamic and plastic cell type and can be divided into two main groups: classically activated macrophages (M1) and alternatively activated macrophages (M2). M2 group can further be divided into sub-groups M2a, M2b, M2c, and M2d. Typically the M2 cells have anti-inflammatory and regulatory properties and produce many different anti-inflammatory cytokines such as IL-4, IL-33, IL-10, IL-1RA, and TGF-β. M2 cells can also secrete angiogenic and chemotactic factors. These cells can be distinguished based on the different expression levels of various surface proteins and the secretion of different effector molecules.

<span class="mw-page-title-main">Dermal macrophage</span> Skin macrophages used for wound repair and hair growth

Dermal macrophages are macrophages in the skin that facilitate skin homeostasis by mediating wound repair, hair growth, and salt balance. Their functional role in these processes is the mediator of inflammation. They can acquire an M1 or M2 phenotype to promote or suppress an inflammatory response, thereby influencing other cells' activity via the production of pro-inflammatory or anti-inflammatory cytokines. Dermal macrophages' ability to acquire pro-inflammatory properties also potentiates them in cancer defence. M1 macrophages can suppress tumour growth in the skin by their pro-inflammatory properties. However, M2 macrophages support tumour growth and invasion by the production of Th2 cytokines such as TGFβ and IL-10. Thus, the exact contribution of each phenotype to cancer defence and the skin's homeostasis is still unclear.

References

  1. Weinberger SE, Cockrill BA, Mandel J (2019). Principles of pulmonary medicine (Seventh ed.). Philadelphia, PA. pp. 288–289. ISBN   978-0-323-52373-8. OCLC   1020498796.{{cite book}}: CS1 maint: location missing publisher (link)
  2. Cotran RS, Kumar V, Collins T, Robbins SL (1999). Robbins Pathologic Basis of Disease. Philadelphia: W.B Saunders Company. ISBN   978-0-7216-7335-6.
  3. Morgan WK (November 1978). "Industrial bronchitis". Br J Ind Med. 35 (4): 285–91. doi:10.1136/oem.35.4.285. PMC   1008445 . PMID   367424.
  4. Perry W, Konopka K. "Lung - Other interstitial pneumonitis / fibrosis - Respiratory bronchiolitis". Topic Completed: 1 July 2020. Minor changes: 1 July 2020.
  5. Zander, Dani (2018). Pulmonary pathology. Philadelphia, PA: Elsevier. ISBN   978-0-323-39308-9. OCLC   968711140. - VII Acute Lung Injury with Siderophages
  6. Majno G, Joris I (12 August 2004). Cells, Tissues, and Disease : Principles of General Pathology. Oxford University Press. p. 620. ISBN   978-0-19-974892-1. OCLC   76950968 . Retrieved 19 March 2013.
  7. 1 2 3 4 Lambrecht, BN (April 2006). ""Alveolar macrophage in the driver's seat"". Immunity. 24 (4): 366–8. doi: 10.1016/j.immuni.2006.03.008 . PMID   16618595.
  8. 1 2 Guyton AC (2007). "Chapter 33: Physiology of the respiratory system". Textbook of Medical Physiology. pp. 431–433.
  9. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 Stafford JL, Neumann NF, Belosevic M (2002). "Macrophage-mediated innate host defense against protozoan parasites". Critical Reviews in Microbiology. 28 (3): 187–248. doi:10.1080/1040-840291046731. PMID   12385499. S2CID   38166749.
  10. 1 2 3 Krutzik SR, Modlin RL (February 2004). "The role of Toll-like receptors in combating mycobacteria". Seminars in Immunology. 16 (1): 35–41. doi:10.1016/j.smim.2003.10.005. PMID   14751762.
  11. 1 2 3 4 5 6 7 8 9 Serezani CH, Chung J, Ballinger MN, Moore BB, Aronoff DM, Peters-Golden M (November 2007). "Prostaglandin E2 suppresses bacterial killing in alveolar macrophages by inhibiting NADPH oxidase". American Journal of Respiratory Cell and Molecular Biology. 37 (5): 562–70. doi:10.1165/rcmb.2007-0153OC. PMC   2048683 . PMID   17585108.
  12. 1 2 Holt PG, Oliver J, Bilyk N, McMenamin C, McMenamin PG, Kraal G, Thepen T (February 1993). "Downregulation of the antigen presenting cell function(s) of pulmonary dendritic cells in vivo by resident alveolar macrophages". The Journal of Experimental Medicine. 177 (2): 397–407. doi:10.1084/jem.177.2.397. PMC   2190916 . PMID   8426110.
  13. 1 2 Bunn HJ, Hewitt CR, Grigg J (May 2002). "Suppression of autologous peripheral blood mononuclear cell proliferation by alveolar macrophages from young infants". Clinical and Experimental Immunology. 128 (2): 313–7. doi:10.1046/j.1365-2249.2002.01848.x. PMC   1906398 . PMID   12041510.
  14. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 Bingisser RM, Holt PG (April 2001). "Immunomodulating mechanisms in the lower respiratory tract: nitric oxide mediated interactions between alveolar macrophages, epithelial cells, and T-cells". Swiss Medical Weekly. 131 (13–14): 171–9. doi:10.4414/smw.2001.09653. PMID   11345807. S2CID   37202272.
  15. 1 2 3 4 5 Lacraz S, Nicod L, Galve-de Rochemonteix B, Baumberger C, Dayer JM, Welgus HG (August 1992). "Suppression of metalloproteinase biosynthesis in human alveolar macrophages by interleukin-4". The Journal of Clinical Investigation. 90 (2): 382–8. doi:10.1172/JCI115872. PMC   443112 . PMID   1322938.
  16. 1 2 3 Brock TG, Serezani CH, Carstens JK, Peters-Golden M, Aronoff DM (January 2008). "Effects of prostaglandin E2 on the subcellular localization of Epac-1 and Rap1 proteins during Fcgamma-receptor-mediated phagocytosis in alveolar macrophages". Experimental Cell Research. 314 (2): 255–63. doi:10.1016/j.yexcr.2007.10.011. PMC   2390918 . PMID   18021770.
  17. Pouliot P, Turmel V, Gélinas E, Laviolette M, Bissonnette EY (June 2005). "Interleukin-4 production by human alveolar macrophages". Clinical and Experimental Allergy. 35 (6): 804–10. doi:10.1111/j.1365-2222.2005.02246.x. PMID   15969673. S2CID   22847451.
  18. 1 2 Paul WE (May 1991). "Interleukin-4: a prototypic immunoregulatory lymphokine". Blood. 77 (9): 1859–70. doi: 10.1182/blood.V77.9.1859.1859 . PMID   2018830.
  19. 1 2 3 Yoshizawa S, Tateda K, Matsumoto T, Gondaira F, Miyazaki S, Standiford TJ, Yamaguchi K (May 2005). "Legionella pneumophila evades gamma interferon-mediated growth suppression through interleukin-10 induction in bone marrow-derived macrophages". Infection and Immunity. 73 (5): 2709–17. doi:10.1128/IAI.73.5.2709-2717.2005. PMC   1087334 . PMID   15845473.
  20. 1 2 Araya J, Cambier S, Morris A, Finkbeiner W, Nishimura SL (August 2006). "Integrin-mediated transforming growth factor-beta activation regulates homeostasis of the pulmonary epithelial-mesenchymal trophic unit". The American Journal of Pathology. 169 (2): 405–15. doi:10.2353/ajpath.2006.060049. PMC   1698780 . PMID   16877343.
  21. Morris DG, Huang X, Kaminski N, Wang Y, Shapiro SD, Dolganov G, Glick A, Sheppard D (March 2003). "Loss of integrin alpha(v)beta6-mediated TGF-beta activation causes Mmp12-dependent emphysema". Nature. 422 (6928): 169–73. Bibcode:2003Natur.422..169M. doi:10.1038/nature01413. PMID   12634787. S2CID   4407206.
  22. 1 2 3 4 5 6 7 8 9 10 11 Takabayshi K, Corr M, Hayashi T, Redecke V, Beck L, Guiney D, Sheppard D, Raz E (April 2006). "Induction of a homeostatic circuit in lung tissue by microbial compounds". Immunity. 24 (4): 475–87. doi: 10.1016/j.immuni.2006.02.008 . PMID   16618605.
  23. 1 2 3 Ray CA, Lasbury ME, Durant PJ, Wang SH, Zhang C, Liao CP, Tschang D, Lee CH (2006). "Transforming growth factor-beta activation and signaling in the alveolar environment during Pneumocystis pneumonia". The Journal of Eukaryotic Microbiology. 53 (Suppl 1): S127–9. doi:10.1111/j.1550-7408.2006.00200.x. PMID   17169028. S2CID   37439751.
  24. 1 2 Annes JP, Munger JS, Rifkin DB (January 2003). "Making sense of latent TGFbeta activation". Journal of Cell Science. 116 (Pt 2): 217–24. doi: 10.1242/jcs.00229 . PMID   12482908.
  25. Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton SL, Wu J, Pittet JF, Kaminski N, Garat C, Matthay MA, Rifkin DB, Sheppard D (February 1999). "The integrin alpha v beta 6 binds and activates latent TGF beta 1: a mechanism for regulating pulmonary inflammation and fibrosis". Cell. 96 (3): 319–28. doi: 10.1016/S0092-8674(00)80545-0 . PMID   10025398.
  26. 1 2 Ohbayashi H, Shimokata K (April 2005). "Matrix metalloproteinase-9 and airway remodeling in asthma". Current Drug Targets. Inflammation and Allergy. 4 (2): 177–81. doi:10.2174/1568010053586246. PMID   15853739.