Serum amyloid A1

Last updated
SAA1
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases SAA1 , PIG4, SAA, SAA2, TP53I4, Serum amyloid A1
External IDs OMIM: 104750 MGI: 98222 HomoloGene: 128033 GeneCards: SAA1
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_199161
NM_000331
NM_001178006

NM_011314
NM_001357491
NM_001379268
NM_001379269

RefSeq (protein)

NP_000322
NP_001171477
NP_954630

NP_035444
NP_001344420
NP_001366197
NP_001366198

Location (UCSC) Chr 11: 18.27 – 18.27 Mb Chr 7: 46.4 – 46.4 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Serum amyloid A1 (SAA1) is a protein that in humans is encoded by the SAA1 gene. [5] [6] [7] SAA1 is a major acute-phase protein mainly produced by hepatocytes in response to infection, tissue injury and malignancy. [8] When released into blood circulation, SAA1 is present as an apolipoprotein associated with high-density lipoprotein (HDL). [9] SAA1 is a major precursor of amyloid A (AA), the deposit of which leads to inflammatory amyloidosis. [10] [11]

Contents

Gene

The gene coding for human SAA1 is one of the 4 SAA genes mapped to a region in the short arm on Chromosome 11. [12] Two of these genes, SAA1 and SAA2, are inducible during acute-phase response, whereas SAA3 is a pseudogene in humans [13] and SAA4 is constitutively expressed in a variety of tissues and cells. Single nucleotide polymorphisms (SNPs) are found in SAA1 in both coding and non-coding sequences, with those located in the coding sequence defining 5 isoforms of SAA1 (SAA1.1 – 1.5). Genetic studies have shown association of some of these SNPs with the disposition to several human diseases including familiar Mediterranean fever, coronary artery diseases, cerebral infarction, and osteoporosis. Mice also have 4 Saa genes. A major difference between human and mouse SAA genes is the expression of the mouse Saa3 gene for a functional protein, generally considered an inducible SAA in inflammatory tissues.

Protein structure

The product of human SAA1 is a pre-protein of 122 amino acids, with a cleavable signal peptide of 18 amino acids. Mature SAA1 consists of 104 amino acids with an apparent molecular weight of 12,500. A crystal structure of SAA1.1 has been solved recently (Figure 1). Native SAA1 is a hexamer with each subunit assuming an antiparallel 4-helix bundle structure. [14] The structure is cone-shaped with its apex forming a binding site for HDL and heparin. The N-terminal helices 1 and 3 have been identified as amyloidogenic peptides of SAA1.1, that are not presence on protein surface in native SAA1 protein. These findings provide the structural basis for the formation of amyloid A fibrils. The human SAA1.1 is comparable at the subunit level with the recently solved structure of mouse Saa3. [15]

Inducible expression

SAA1 and SAA2 are highly inducible and hence called acute-phase SAA. Inflammatory cytokines such as IL-1β, IL-6 and TNF-α are major stimulants for hepatocyte expression of the SAA1 gene. [16] Inducible expression of the acute-phase SAA genes is mainly regulated at the transcription level and involves the transcription factors C/EBP, NF-κB, AP2, SAF, Sp1 and STAT3. Elevation of the transcript of SAA1 is often seen in cDNA arrays used for detection of proinflammatory cytokine expression. SAA1 protein level correlates with its transcript level, and has long been considered a clinical indicator for inflammatory conditions.

Interactions

In addition to its association with HDL, SAA1 interacts with a number of mammalian proteins, mostly cell surface proteins such as receptors. SAA1 binding to the αvβ3 integrin produces an inhibitory effect on the growth of nasopharyngeal carcinoma. [17] Several receptors for SAA1 have been identified using an SAA1 hybrid protein containing two amino acid substitutions from SAA2. [18] These receptors include the G protein-coupled chemoattractant receptor FPR2 (formyl peptide receptor 2), [19] believed to mediate the chemotactic activity of the recombinant SAA1; the murine scavenger receptor SR-BI [20] and the human equivalent CLA-1., [21] for a possible role in SAA1-dependent cholesterol metabolism. Moreover, the Toll-like receptors TLR2 [22] and TLR4 [23] mediate SAA1-induced cytokine gene expression. The P2X7 purinergic receptor is another receptor used by SAA1 for a number of cellular functions including the activation of NLRP3 inflammasomes. [24]

SAA1 has been found to interact with outer membrane protein A (ompA) of several Gram-negative bacteria including E. coli, Salmonella typhimurium , Shigella flexneri , Vibrio cholerae and P. aeruginosa. [25] Exposure of these Gram-negative bacteria to SAA1 promotes uptake of the bacteria by neutrophils, suggesting that SAA1 serves as an opsonin that enhances bacteria clearance. [26] A more recent study identified SAA1 interaction with retinol, resulting in reduced bacterial burden. [27] These findings suggest that SAA1 has a function in host defense against bacterial infection.

Functions and clinical relevance

The biological function of SAA1 has not been fully understood despite intensive research in the last three decades. Research tools such as the SAA1 knockout mice and transgenic mice have become available only recently. It has been well established, however, that elevated plasma concentration of SAA1 is associated with a multitude of inflammatory conditions. As a result, SAA1 has been a clinical indicator and reliable biomarker for inflammatory diseases, chronic metabolic disorders and late-stage malignancy. [28] Inflammatory amyloidosis results from chronic inflammation with increased production of SAA1, which is a major precursor of amyloid A fibril deposit in various tissues. [29]

SAA1 has been extensively studied for its binding to HDL, with results suggesting a role in lipid metabolism. During the acute-phase response, elevated levels of SAA1 in the plasma displaces ApoA-I and becomes a major apolipoprotein of HDL. [30] The exact biological consequence of HDL remodeling by SAA1 is still under investigation, using recently developed tools such as the Saa1 and Saa2 knockout mice. SAA1 is also believed to contribute to the development of atherosclerosis. [31] [32] However, in an ApoE-deficient mouse model, deletion of the Saa1/Saa2 genes does not appear to affect atherosclerotic lesions. [33]

Ex vivo and in vitro studies have shown that the recombinant human SAA1 hybrid protein has strong chemotactic activity for neutrophils and macrophages. [34] This effect is believed to be mediated through FPR2, a G protein-coupled chemoattractant receptor. [35] The same receptor also mediates the cytokine-like activity of the recombinant SAA1, resulting in an elevated expression of IL-8 in neutrophils. [36] The recombinant SAA1 has been reported to induce the expression of a variety of inflammatory cytokines including IL-1β, TNF-α, IL-6, IL-12p40, [37] [38] as well as immunoregulatory cytokines such as IL-23, [39] IL-33 [40] and growth-stimulatory cytokines such as G-CSF. [41]

SAA1 may also be produced by macrophages and epithelial cells in various tissues. It has been shown to promote local Th17 response in the gut. [42] This finding, which is based on both Saa1/Saa2 knockout mice and ex vivo studies of T cells, strongly suggest a local immunomodulatory function of SAA1 as opposed to its established role as an acute-phase protein produced in the liver and present in the plasma as an apolipoprotein of HDL. Transgenic expression of human SAA1.1 in mouse liver aggravates T cell-mediated hepatitis through elevated production of chemokines, [43] which involves the SAA1 receptor TLR2. Secretion of SAA1 by melanoma cells may induce anti-inflammatory IL-10-secreting neutrophils that interact with invariant natural killer T cells (iNKT cells). [44] In addition, SAA1 can skew macrophages to a M2 phenotype. [45]

Published reports link SAA1 to a number of malignancies, but a causal relationship has not been established. SAA1 has been associated with tumor pathogenesis, [46] and its gene polymorphism is a contributing factor to certain types of malignant tumors. [47] SAA1 has also been shown to affect the tumor microenvironment and contribute to tumor cell metastasis. [48]

Some of the results obtained with the recombinant human SAA1 hybrid remain controversial, as the protein does not have exactly the same sequence of human SAA1 and its properties may be different from the native SAA1. [49] Other studies have shown that native human SAA1 retains some of the cytokine-like activities such as the G-CSF-induction capability [50]

Recent studies using the Saa1/Saa2 knockout mice showed weakened Th17 response in gut epithelial cells, [51] suggesting that SAA1 plays a role in vivo in the regulation of immunity.

Notes

Related Research Articles

<span class="mw-page-title-main">Acute-phase protein</span> Class of proteins involved in inflammation

Acute-phase proteins (APPs) are a class of proteins whose concentrations in blood plasma either increase or decrease in response to inflammation. This response is called the acute-phase reaction. The acute-phase reaction characteristically involves fever, acceleration of peripheral leukocytes, circulating neutrophils and their precursors. The terms acute-phase protein and acute-phase reactant (APR) are often used synonymously, although some APRs are polypeptides rather than proteins.

Interleukins (ILs) are a group of cytokines that are expressed and secreted by white blood cells (leukocytes) as well as some other body cells. The human genome encodes more than 50 interleukins and related proteins.

<span class="mw-page-title-main">Lipoxin</span> Acronym for lipoxygenase interaction product

A lipoxin (LX or Lx), an acronym for lipoxygenase interaction product, is a bioactive autacoid metabolite of arachidonic acid made by various cell types. They are categorized as nonclassic eicosanoids and members of the specialized pro-resolving mediators (SPMs) family of polyunsaturated fatty acid (PUFA) metabolites. Like other SPMs, LXs form during, and then act to resolve, inflammatory responses. Initially, two lipoxins were identified, lipoxin A4 (LXA4) and LXB4, but more recent studies have identified epimers of these two LXs: the epi-lipoxins, 15-epi-LXA4 and 15-epi-LXB4 respectively.

<span class="mw-page-title-main">Interleukin 8</span> Mammalian protein found in Homo sapiens

Interleukin 8 is a chemokine produced by macrophages and other cell types such as epithelial cells, airway smooth muscle cells and endothelial cells. Endothelial cells store IL-8 in their storage vesicles, the Weibel-Palade bodies. In humans, the interleukin-8 protein is encoded by the CXCL8 gene. IL-8 is initially produced as a precursor peptide of 99 amino acids which then undergoes cleavage to create several active IL-8 isoforms. In culture, a 72 amino acid peptide is the major form secreted by macrophages.

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

Interleukin-1 alpha also known as hematopoietin 1 is a cytokine of the interleukin 1 family that in humans is encoded by the IL1A gene. In general, Interleukin 1 is responsible for the production of inflammation, as well as the promotion of fever and sepsis. IL-1α inhibitors are being developed to interrupt those processes and treat diseases.

<span class="mw-page-title-main">Interleukin 15</span> Cytokine with structural similarity to Interleukin-2

Interleukin-15 (IL-15) is a protein that in humans is encoded by the IL15 gene. IL-15 is an inflammatory cytokine with structural similarity to Interleukin-2 (IL-2). Like IL-2, IL-15 binds to and signals through a complex composed of IL-2/IL-15 receptor beta chain (CD122) and the common gamma chain. IL-15 is secreted by mononuclear phagocytes following infection by virus(es). This cytokine induces the proliferation of natural killer cells, i.e. cells of the innate immune system whose principal role is to kill virally infected cells.

<span class="mw-page-title-main">Interleukin 18</span> Protein-coding gene in the species Homo sapiens

Interleukin-18 (IL-18), also known as interferon-gamma inducing factor is a protein which in humans is encoded by the IL18 gene. The protein encoded by this gene is a proinflammatory cytokine. Many cell types, both hematopoietic cells and non-hematopoietic cells, have the potential to produce IL-18. It was first described in 1989 as a factor that induced interferon-γ (IFN-γ) production in mouse spleen cells. Originally, IL-18 production was recognized in Kupffer cells, liver-resident macrophages. However, IL-18 is constitutively expressed in non-hematopoietic cells, such as intestinal epithelial cells, keratinocytes, and endothelial cells. IL-18 can modulate both innate and adaptive immunity and its dysregulation can cause autoimmune or inflammatory diseases.

<span class="mw-page-title-main">Interleukin 33</span> IL-33 induces helper T cells, mast cells, eosinophils and basophils to produce type 2 cytokines.

Interleukin 33 (IL-33) is a protein that in humans is encoded by the IL33 gene.

<span class="mw-page-title-main">Interleukin 26</span>

Interleukin-26 (IL-26) is a protein that in humans is encoded by the IL26 gene.

<span class="mw-page-title-main">Interleukin 22</span>

Interleukin-22 (IL-22) is protein that in humans is encoded by the IL22 gene.

<span class="mw-page-title-main">Interleukin 17</span> Group of proteins

Interleukin 17 family is a family of pro-inflammatory cystine knot cytokines. They are produced by a group of T helper cell known as T helper 17 cell in response to their stimulation with IL-23. Originally, Th17 was identified in 1993 by Rouvier et al. who isolated IL17A transcript from a rodent T-cell hybridoma. The protein encoded by IL17A is a founding member of IL-17 family. IL17A protein exhibits a high homology with a viral IL-17-like protein encoded in the genome of T-lymphotropic rhadinovirus Herpesvirus saimiri. In rodents, IL-17A is often referred to as CTLA8.

Serum amyloid A (SAA) proteins are a family of apolipoproteins associated with high-density lipoprotein (HDL) in plasma. Different isoforms of SAA are expressed constitutively at different levels or in response to inflammatory stimuli. These proteins are produced predominantly by the liver.

Chemokine ligands 4 previously known as macrophage inflammatory protein (MIP-1β), is a protein which in humans is encoded by the CCL4 gene. CCL4 belongs to a cluster of genes located on 17q11-q21 of the chromosomal region. Identification and localization of the gene on the chromosome 17 was in 1990 although the discovery of MIP-1 was initiated in 1988 with the purification of a protein doublet corresponding to inflammatory activity from supernatant of endotoxin-stimulated murine macrophages. At that time, it was also named as "macrophage inflammatory protein-1" (MIP-1) due to its inflammatory properties.

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

CCAAT/enhancer-binding protein beta is a protein that in humans is encoded by the CEBPB gene.

<span class="mw-page-title-main">PTX3</span>

Pentraxin-related protein PTX3 also known as TNF-inducible gene 14 protein (TSG-14) is a protein that in humans is encoded by the PTX3 gene.

<span class="mw-page-title-main">Formyl peptide receptor 2</span> Protein-coding gene in the species Homo sapiens

N-formyl peptide receptor 2 (FPR2) is a G-protein coupled receptor (GPCR) located on the surface of many cell types of various animal species. The human receptor protein is encoded by the FPR2 gene and is activated to regulate cell function by binding any one of a wide variety of ligands including not only certain N-Formylmethionine-containing oligopeptides such as N-Formylmethionine-leucyl-phenylalanine (FMLP) but also the polyunsaturated fatty acid metabolite of arachidonic acid, lipoxin A4 (LXA4). Because of its interaction with lipoxin A4, FPR2 is also commonly named the ALX/FPR2 or just ALX receptor.

<span class="mw-page-title-main">SAA2</span> Protein-coding gene in the species Homo sapiens

Serum amyloid A protein is a protein that in humans is encoded by the SAA2 gene.

<span class="mw-page-title-main">Interleukin 37</span> Protein-coding gene in the species Homo sapiens

Interleukin 37 (IL-37), also known as Interleukin-1 family member 7 (IL-1F7), is an anti-inflammatory cytokine important for the downregulation of pro-inflammatory cytokine production as well as the suppression of tumor cell growth.

<span class="mw-page-title-main">Interleukin-1 family</span> Group of cytokines playing a key role in the regulation of immune and inflammatory responses

The Interleukin-1 family is a group of 11 cytokines that play a central role in the regulation of immune and inflammatory responses to infections or sterile insults.

Interleukin 36, or IL-36, is a group of cytokines in the IL-1 family with pro-inflammatory effects. The role of IL-36 in inflammatory diseases is under investigation.

References

  1. 1 2 3 ENSG00000288411 GRCh38: Ensembl release 89: ENSG00000173432, ENSG00000288411 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000057465 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. Glaser T, Housman D, Lewis WH, Gerhard D, Jones C (Nov 1989). "A fine-structure deletion map of human chromosome 11p: analysis of J1 series hybrids". Somatic Cell and Molecular Genetics. 15 (6): 477–501. doi:10.1007/BF01534910. PMID   2595451. S2CID   19607294.
  6. Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B (Sep 1997). "A model for p53-induced apoptosis". Nature. 389 (6648): 300–5. doi:10.1038/38525. PMID   9305847. S2CID   4429638.
  7. "Entrez Gene: SAA1 serum amyloid A1".
  8. Gabay C, Kushner I (Feb 1999). "Acute-phase proteins and other systemic responses to inflammation". The New England Journal of Medicine. 340 (6): 448–54. doi:10.1056/NEJM199902113400607. PMID   9971870.
  9. Benditt EP, Eriksen N (Sep 1977). "Amyloid protein SAA is associated with high density lipoprotein from human serum". Proceedings of the National Academy of Sciences of the United States of America. 74 (9): 4025–8. doi: 10.1073/pnas.74.9.4025 . PMC   431828 . PMID   198813.
  10. Husebekk A, Skogen B, Husby G, Marhaug G (Mar 1985). "Transformation of amyloid precursor SAA to protein AA and incorporation in amyloid fibrils in vivo". Scandinavian Journal of Immunology. 21 (3): 283–7. doi:10.1111/j.1365-3083.1985.tb01431.x. PMID   3922050. S2CID   29482438.
  11. Tape C, Tan R, Nesheim M, Kisilevsky R (Sep 1988). "Direct evidence for circulating apoSAA as the precursor of tissue AA amyloid deposits". Scandinavian Journal of Immunology. 28 (3): 317–24. doi:10.1111/j.1365-3083.1988.tb01455.x. PMID   3194701. S2CID   35274998.
  12. Uhlar CM, Burgess CJ, Sharp PM, Whitehead AS (Jan 1994). "Evolution of the serum amyloid A (SAA) protein superfamily". Genomics. 19 (2): 228–35. doi:10.1006/geno.1994.1052. PMID   8188253.
  13. Kluve-Beckerman B, Drumm ML, Benson MD (Nov 1991). "Nonexpression of the human serum amyloid A three (SAA3) gene". DNA and Cell Biology. 10 (9): 651–61. doi:10.1089/dna.1991.10.651. PMID   1755958.
  14. Lu J, Yu Y, Zhu I, Cheng Y, Sun PD (Apr 2014). "Structural mechanism of serum amyloid A-mediated inflammatory amyloidosis". Proceedings of the National Academy of Sciences of the United States of America. 111 (14): 5189–94. doi: 10.1073/pnas.1322357111 . PMC   3986191 . PMID   24706838.
  15. Derebe MG, Zlatkov CM, Gattu S, Ruhn KA, Vaishnava S, Diehl GE, MacMillan JB, Williams NS, Hooper LV (Jul 2014). "Serum amyloid A is a retinol binding protein that transports retinol during bacterial infection". eLife. 3: e03206. doi:10.7554/eLife.03206. PMC   4129439 . PMID   25073702.
  16. Jensen LE, Whitehead AS (Sep 1998). "Regulation of serum amyloid A protein expression during the acute-phase response". The Biochemical Journal. 334 (3): 489–503. doi:10.1042/bj3340489. PMC   1219714 . PMID   9729453.
  17. Lung HL, Man OY, Yeung MC, Ko JM, Cheung AK, Law EW, Yu Z, Shuen WH, Tung E, Chan SH, Bangarusamy DK, Cheng Y, Yang X, Kan R, Phoon Y, Chan KC, Chua D, Kwong DL, Lee AW, Ji MF, Lung ML (Feb 2015). "SAA1 polymorphisms are associated with variation in antiangiogenic and tumor-suppressive activities in nasopharyngeal carcinoma". Oncogene. 34 (7): 878–89. doi: 10.1038/onc.2014.12 . PMID   24608426. S2CID   9519362.
  18. "Recombinant human Apo-SAA".[ permanent dead link ]
  19. Su SB, Gong W, Gao JL, Shen W, Murphy PM, Oppenheim JJ, Wang JM (Jan 1999). "A seven-transmembrane, G protein-coupled receptor, FPRL1, mediates the chemotactic activity of serum amyloid A for human phagocytic cells". The Journal of Experimental Medicine. 189 (2): 395–402. doi:10.1084/jem.189.2.395. PMC   2192984 . PMID   9892621.
  20. Cai L, de Beer MC, de Beer FC, van der Westhuyzen DR (Jan 2005). "Serum amyloid A is a ligand for scavenger receptor class B type I and inhibits high density lipoprotein binding and selective lipid uptake". The Journal of Biological Chemistry. 280 (4): 2954–61. doi: 10.1074/jbc.M411555200 . PMID   15561721.
  21. Baranova IN, Vishnyakova TG, Bocharov AV, Kurlander R, Chen Z, Kimelman ML, Remaley AT, Csako G, Thomas F, Eggerman TL, Patterson AP (Mar 2005). "Serum amyloid A binding to CLA-1 (CD36 and LIMPII analogous-1) mediates serum amyloid A protein-induced activation of ERK1/2 and p38 mitogen-activated protein kinases". The Journal of Biological Chemistry. 280 (9): 8031–40. doi: 10.1074/jbc.M405009200 . PMID   15576377.
  22. Cheng N, He R, Tian J, Ye PP, Ye RD (Jul 2008). "Cutting edge: TLR2 is a functional receptor for acute-phase serum amyloid A". Journal of Immunology. 181 (1): 22–6. doi:10.4049/jimmunol.181.1.22. PMC   2464454 . PMID   18566366.
  23. Sandri S, Rodriguez D, Gomes E, Monteiro HP, Russo M, Campa A (May 2008). "Is serum amyloid A an endogenous TLR4 agonist?". Journal of Leukocyte Biology. 83 (5): 1174–80. doi: 10.1189/jlb.0407203 . PMID   18252871. S2CID   17352286.
  24. Niemi K, Teirilä L, Lappalainen J, Rajamäki K, Baumann MH, Öörni K, Wolff H, Kovanen PT, Matikainen S, Eklund KK (Jun 2011). "Serum amyloid A activates the NLRP3 inflammasome via P2X7 receptor and a cathepsin B-sensitive pathway". Journal of Immunology. 186 (11): 6119–28. doi: 10.4049/jimmunol.1002843 . PMID   21508263.
  25. Hari-Dass R, Shah C, Meyer DJ, Raynes JG (May 2005). "Serum amyloid A protein binds to outer membrane protein A of gram-negative bacteria". The Journal of Biological Chemistry. 280 (19): 18562–7. doi: 10.1074/jbc.M500490200 . PMID   15705572.
  26. Shah C, Hari-Dass R, Raynes JG (Sep 2006). "Serum amyloid A is an innate immune opsonin for Gram-negative bacteria". Blood. 108 (5): 1751–7. doi: 10.1182/blood-2005-11-011932 . PMID   16735604.
  27. Derebe MG, Zlatkov CM, Gattu S, Ruhn KA, Vaishnava S, Diehl GE, MacMillan JB, Williams NS, Hooper LV (29 July 2014). "Serum amyloid A is a retinol binding protein that transports retinol during bacterial infection". eLife. 3: e03206. doi:10.7554/eLife.03206. PMC   4129439 . PMID   25073702.
  28. Malle E, Sodin-Semrl S, Kovacevic A (Jan 2009). "Serum amyloid A: an acute-phase protein involved in tumour pathogenesis". Cellular and Molecular Life Sciences. 66 (1): 9–26. doi:10.1007/s00018-008-8321-x. PMC   4864400 . PMID   18726069.
  29. Tape C, Tan R, Nesheim M, Kisilevsky R (Sep 1988). "Direct evidence for circulating apoSAA as the precursor of tissue AA amyloid deposits". Scandinavian Journal of Immunology. 28 (3): 317–24. doi:10.1111/j.1365-3083.1988.tb01455.x. PMID   3194701. S2CID   35274998.
  30. Kisilevsky R, Manley PN (Mar 2012). "Acute-phase serum amyloid A: perspectives on its physiological and pathological roles". Amyloid. 19 (1): 5–14. doi:10.3109/13506129.2011.654294. PMID   22320226. S2CID   43235598.
  31. King VL, Thompson J, Tannock LR (Aug 2011). "Serum amyloid A in atherosclerosis". Current Opinion in Lipidology. 22 (4): 302–7. doi:10.1097/MOL.0b013e3283488c39. PMID   21734573. S2CID   10073239.
  32. Thompson JC, Jayne C, Thompson J, Wilson PG, Yoder MH, Webb N, Tannock LR (Feb 2015). "A brief elevation of serum amyloid A is sufficient to increase atherosclerosis". Journal of Lipid Research. 56 (2): 286–93. doi:10.1194/jlr.M054015. PMC   4306683 . PMID   25429103.
  33. De Beer MC, Wroblewski JM, Noffsinger VP, Rateri DL, Howatt DA, Balakrishnan A, Ji A, Shridas P, Thompson JC, van der Westhuyzen DR, Tannock LR, Daugherty A, Webb NR, De Beer FC (Feb 2014). "Deficiency of endogenous acute phase serum amyloid A does not affect atherosclerotic lesions in apolipoprotein E-deficient mice". Arteriosclerosis, Thrombosis, and Vascular Biology. 34 (2): 255–61. doi:10.1161/ATVBAHA.113.302247. PMC   3951741 . PMID   24265416.
  34. Badolato R, Wang JM, Murphy WJ, Lloyd AR, Michiel DF, Bausserman LL, Kelvin DJ, Oppenheim JJ (Jul 1994). "Serum amyloid A is a chemoattractant: induction of migration, adhesion, and tissue infiltration of monocytes and polymorphonuclear leukocytes". The Journal of Experimental Medicine. 180 (1): 203–9. doi:10.1084/jem.180.1.203. PMC   2191543 . PMID   7516407.
  35. Su SB, Gong W, Gao JL, Shen W, Murphy PM, Oppenheim JJ, Wang JM (Jan 1999). "A seven-transmembrane, G protein-coupled receptor, FPRL1, mediates the chemotactic activity of serum amyloid A for human phagocytic cells". The Journal of Experimental Medicine. 189 (2): 395–402. doi:10.1084/jem.189.2.395. PMC   2192984 . PMID   9892621.
  36. He R, Sang H, Ye RD (Feb 2003). "Serum amyloid A induces IL-8 secretion through a G protein-coupled receptor, FPRL1/LXA4R". Blood. 101 (4): 1572–81. doi: 10.1182/blood-2002-05-1431 . PMID   12393391.
  37. Patel H, Fellowes R, Coade S, Woo P (Oct 1998). "Human serum amyloid A has cytokine-like properties". Scandinavian Journal of Immunology. 48 (4): 410–8. doi: 10.1046/j.1365-3083.1998.00394.x . PMID   9790312. S2CID   44779960.
  38. Cheng N, He R, Tian J, Ye PP, Ye RD (Jul 2008). "Cutting edge: TLR2 is a functional receptor for acute-phase serum amyloid A". Journal of Immunology. 181 (1): 22–6. doi:10.4049/jimmunol.181.1.22. PMC   2464454 . PMID   18566366.
  39. He R, Shepard LW, Chen J, Pan ZK, Ye RD (Sep 2006). "Serum amyloid A is an endogenous ligand that differentially induces IL-12 and IL-23". Journal of Immunology. 177 (6): 4072–9. doi: 10.4049/jimmunol.177.6.4072 . PMID   16951371.
  40. Sun L, Zhu Z, Cheng N, Yan Q, Ye RD (Jul 2014). "Serum amyloid A induces interleukin-33 expression through an IRF7-dependent pathway". European Journal of Immunology. 44 (7): 2153–64. doi:10.1002/eji.201344310. PMC   4118754 . PMID   24777946.
  41. He RL, Zhou J, Hanson CZ, Chen J, Cheng N, Ye RD (Jan 2009). "Serum amyloid A induces G-CSF expression and neutrophilia via Toll-like receptor 2". Blood. 113 (2): 429–37. doi:10.1182/blood-2008-03-139923. PMC   2615655 . PMID   18952897.
  42. Sano T, Huang W, Hall JA, Yang Y, Chen A, Gavzy SJ, Lee JY, Ziel JW, Miraldi ER, Domingos AI, Bonneau R, Littman DR (Oct 2015). "An IL-23R/IL-22 Circuit Regulates Epithelial Serum Amyloid A to Promote Local Effector Th17 Responses". Cell. 163 (2): 381–93. doi:10.1016/j.cell.2015.08.061. PMC   4621768 . PMID   26411290.
  43. Ji YR, Kim HJ, Bae KB, Lee S, Kim MO, Ryoo ZY (May 2015). "Hepatic serum amyloid A1 aggravates T cell-mediated hepatitis by inducing chemokines via Toll-like receptor 2 in mice". The Journal of Biological Chemistry. 290 (20): 12804–11. doi: 10.1074/jbc.M114.635763 . PMC   4432296 . PMID   25847238.
  44. De Santo C, Arscott R, Booth S, Karydis I, Jones M, Asher R, Salio M, Middleton M, Cerundolo V (Nov 2010). "Invariant NKT cells modulate the suppressive activity of IL-10-secreting neutrophils differentiated with serum amyloid A". Nature Immunology. 11 (11): 1039–46. doi:10.1038/ni.1942. PMC   3001335 . PMID   20890286.
  45. Sun L, Zhou H, Zhu Z, Yan Q, Wang L, Liang Q, Ye RD (May 2015). "Ex vivo and in vitro effect of serum amyloid a in the induction of macrophage M2 markers and efferocytosis of apoptotic neutrophils". Journal of Immunology. 194 (10): 4891–900. doi:10.4049/jimmunol.1402164. PMC   4417396 . PMID   25870242.
  46. Malle E, Sodin-Semrl S, Kovacevic A (Jan 2009). "Serum amyloid A: an acute-phase protein involved in tumour pathogenesis". Cellular and Molecular Life Sciences. 66 (1): 9–26. doi:10.1007/s00018-008-8321-x. PMC   4864400 . PMID   18726069.
  47. Lung HL, Man OY, Yeung MC, Ko JM, Cheung AK, Law EW, Yu Z, Shuen WH, Tung E, Chan SH, Bangarusamy DK, Cheng Y, Yang X, Kan R, Phoon Y, Chan KC, Chua D, Kwong DL, Lee AW, Ji MF, Lung ML (Feb 2015). "SAA1 polymorphisms are associated with variation in antiangiogenic and tumor-suppressive activities in nasopharyngeal carcinoma". Oncogene. 34 (7): 878–89. doi: 10.1038/onc.2014.12 . PMID   24608426. S2CID   9519362.
  48. Hansen MT, Forst B, Cremers N, Quagliata L, Ambartsumian N, Grum-Schwensen B, Klingelhöfer J, Abdul-Al A, Herrmann P, Osterland M, Stein U, Nielsen GH, Scherer PE, Lukanidin E, Sleeman JP, Grigorian M (Jan 2015). "A link between inflammation and metastasis: serum amyloid A1 and A3 induce metastasis, and are targets of metastasis-inducing S100A4". Oncogene. 34 (4): 424–35. doi: 10.1038/onc.2013.568 . PMID   24469032. S2CID   5774746.
  49. Christenson K, Björkman L, Ahlin S, Olsson M, Sjöholm K, Karlsson A, Bylund J (2013). "Endogenous Acute Phase Serum Amyloid A Lacks Pro-Inflammatory Activity, Contrasting the Two Recombinant Variants That Activate Human Neutrophils through Different Receptors". Frontiers in Immunology. 4: 92. doi: 10.3389/fimmu.2013.00092 . PMC   3631709 . PMID   23626589.
  50. Kim MH, de Beer MC, Wroblewski JM, Webb NR, de Beer FC (Feb 2013). "SAA does not induce cytokine production in physiological conditions". Cytokine. 61 (2): 506–12. doi:10.1016/j.cyto.2012.10.019. PMC   3616876 . PMID   23165195.
  51. Sano T, Huang W, Hall JA, Yang Y, Chen A, Gavzy SJ, Lee JY, Ziel JW, Miraldi ER, Domingos AI, Bonneau R, Littman DR (Oct 2015). "An IL-23R/IL-22 Circuit Regulates Epithelial Serum Amyloid A to Promote Local Effector Th17 Responses". Cell. 163 (2): 381–93. doi:10.1016/j.cell.2015.08.061. PMC   4621768 . PMID   26411290.

Further reading