Novel Majeed Syndrome-Causing LPIN2 Mutations Link Bone Inflammation to Inflammatory M2 Macrophages and Accelerated Osteoclastogenesis

Farzana Bhuyan, Adriana A de Jesus, Jacob Mitchell, Evgenia Leikina, Rachel VanTries, Ronit Herzog, Karen B Onel, Andrew Oler, Gina A Montealegre Sanchez, Kim A Johnson, Lena Bichell, Bernadette Marrero, Luis Fernandez De Castro, Yan Huang, Katherine R Calvo, Michael T Collins, Sundar Ganesan, Leonid V Chernomordik, Polly J Ferguson, Raphaela Goldbach-Mansky, Farzana Bhuyan, Adriana A de Jesus, Jacob Mitchell, Evgenia Leikina, Rachel VanTries, Ronit Herzog, Karen B Onel, Andrew Oler, Gina A Montealegre Sanchez, Kim A Johnson, Lena Bichell, Bernadette Marrero, Luis Fernandez De Castro, Yan Huang, Katherine R Calvo, Michael T Collins, Sundar Ganesan, Leonid V Chernomordik, Polly J Ferguson, Raphaela Goldbach-Mansky

Abstract

Objective: To identify novel heterozygous LPIN2 mutations in a patient with Majeed syndrome and characterize the pathomechanisms that lead to the development of sterile osteomyelitis.

Methods: Targeted genetic analysis and functional studies assessing monocyte responses, macrophage differentiation, and osteoclastogenesis were conducted to compare the pathogenesis of Majeed syndrome to interleukin-1 (IL-1)-mediated diseases including neonatal-onset multisystem inflammatory disease (NOMID) and deficiency of the IL-1 receptor antagonist (DIRA).

Results: A 4-year-old girl of mixed ethnic background presented with sterile osteomyelitis and elevated acute-phase reactants. She had a 17.8-kb deletion on the maternal LPIN2 allele and a splice site mutation, p.R517H, that variably spliced out exons 10 and 11 on the paternal LPIN2 allele. The patient achieved long-lasting remission receiving IL-1 blockade with canakinumab. Compared to controls, monocytes and monocyte-derived M1-like macrophages from the patient with Majeed syndrome and those with NOMID or DIRA had elevated caspase 1 activity and IL-1β secretion. In contrast, lipopolysaccharide-stimulated, monocyte-derived, M2-like macrophages from the patient with Majeed syndrome released higher levels of osteoclastogenic mediators (IL-8, IL-6, tumor necrosis factor, CCL2, macrophage inflammatory protein 1α/β, CXCL8, and CXCL1) compared to NOMID patients and healthy controls. Accelerated osteoclastogenesis in the patient with Majeed syndrome was associated with higher NFATc1 levels, enhanced JNK/MAPK, and reduced Src kinase activation, and partially responded to JNK inhibition and IL-1 (but not IL-6) blockade.

Conclusion: We report 2 novel compound heterozygous disease-causing mutations in LPIN2 in an American patient with Majeed syndrome. LPIN2 deficiency drives differentiation of proinflammatory M2-like macrophages and enhances intrinsic osteoclastogenesis. This provides a model for the pathogenesis of sterile osteomyelitis which differentiates Majeed syndrome from other IL-1-mediated autoinflammatory diseases.

Trial registration: ClinicalTrials.gov NCT02974595.

© 2020 The Authors. Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology. This article is a U.S. Government work and is in the public domain in the USA.

Figures

Figure 1
Figure 1
Novel mutations in LPIN2 cause a compound heterozygous form of Majeed syndrome. A, The patient with Majeed syndrome was compound heterozygous (Comp Het) for 2 rare variants in LPIN2. B, Magnetic resonance imaging (STIR imaging) of the patient’s knee shows bone marrow enhancement without cortical lesions. C, Genomic structure is shown along with location of the LPIN2 locus and known disease‐causing LPIN2 variants reported in the literature (blue). The patient’s maternal allele is depicted with deleted exons (red), while the paternal allele is depicted with splice site mutations, including spliced out exon 10 (red) or exons 10 and 11 (orange). D, LPIN2 splice junctions were quantified in the patient’s blood using alignment data on whole‐blood RNA‐Seq analysis, yielding predicted transcript levels with each deletion (healthy control [HC] data not shown). Prediction of 56% of the transcript with the maternal deletion suggests that this mutation confers protection against nonsense‐mediated mRNA decay; the 2 alternatively spliced transcripts lacked either exon 10 alone (17.2% of the transcript) or exons 10 and 11 (26.7% of the transcript). E, LPIN2 mRNA expression in human cell subsets and tissues was quantified by real‐time quantitative polymerase chain reaction. Fold changes in LPIN2 expression (relative to 18S) are shown; bars show the mean ± SD of 3 technical replicates. F, Levels of LPIN1, LPIN2, and LPIN3 mRNA were quantified by RNA‐Seq analysis in human monocytes from healthy controls; bars show the mean ± SD of 3 samples. PBMC = peripheral blood mononuclear cell; NK = natural killer; HUVEC = human umbilical vein endothelial cell.
Figure 2
Figure 2
Effect of disease‐causing LPIN2 mutations on interleukin‐1β (IL‐1β) production in monocytes and macrophages. IL‐1β concentration in supernatant was analyzed by enzyme‐linked immunosorbent assay. A, Adherent monocytes from peripheral blood mononuclear cells were stimulated with lipopolysaccharide (LPS; 1 μg/ml) for 2.5 hours and ATP (1 mmole) for 30 minutes. Each symbol represents an individual subject: healthy controls (HCs) (n = 4), patient with Majeed syndrome (n = 1), patients with neonatal‐onset multisystem inflammatory disease (NOMID) (n = 3), and patients with deficiency of the IL‐1 receptor antagonist (DIRA) (n = 3). Bars show the mean ± SD. Values from the patient with Majeed syndrome represent the mean of 3 technical replicates. B, LPS‐ and ATP‐stimulated monocytes were cultured in the presence or absence of anakinra (10 mg/ml). Each symbol represents an individual subject: healthy controls (n = 3) and patient with Majeed syndrome (n = 1). Bars show the mean ± SD. Values from the patient with Majeed syndrome were obtained from 3 independent experiments, using samples from separate visits, with the mean of 2 technical replicates shown. C and D, M1‐like macrophages (C) and M2‐like macrophages (D) were stimulated with LPS (1 μg/ml) and ATP (1 mmole) for 24 hours. Each symbol represents an individual subject: healthy controls (n = 6), patient with Majeed syndrome (n = 1), patients with NOMID (n = 3), and patients with DIRA (n = 4). Bars show the mean ± SD. Values for the patient with Majeed syndrome represent the mean of the technical replicates. * = P < 0.05; ** = P < 0.001, by 2‐sample t‐test assuming equal variances to compare disease groups, or by Student’s unpaired t‐test to compare stimulation with anakinra to no stimulation with anakinra.
Figure 3
Figure 3
Cytokine and chemokine production upon stimulation of M1‐like macrophages (M1‐MDMs) and M2‐MDMs. A and B, M1‐MDMs (A) and M2‐MDMs (B) were stimulated with LPS (1 μg/ml) and ATP (1 mmole) for 24 hours. The relative levels of various cytokines and chemokines in supernatant were analyzed by antibody array, and cytokine density was analyzed using ImageJ software. C, IL‐10 levels were analyzed by enzyme‐linked immunosorbent assay. Each symbol represents an individual subject: healthy controls (n = 4), patient with Majeed syndrome (n = 1), patients with NOMID (n = 3), and patients with DIRA (n = 4). Bars show the mean ± SD. Values from the patient with Majeed syndrome represent the mean of 2 technical replicates.* = P < 0.05; ** = P < 0.001; *** = P < 0.0001, by 2‐sample t‐test assuming equal variances. MCP‐1 = monocyte chemotactic protein 1; GROα = growth‐related oncogene α; TNF = tumor necrosis factor; MIP‐1α/β = macrophage inflammatory protein 1α/β; IP‐10 = interferon‐γ–inducible protein 10 (see Figure 2 for other definitions).
Figure 4
Figure 4
M2‐like macrophages (M2‐MDMs) in Majeed syndrome are more osteoclastogenic, and anakinra could reduce osteoclast fusion. A, MDMs were differentiated with macrophage colony‐stimulating factor (M‐CSF; 100 ng/ml) for 6 days and with a combination of M‐CSF (100 ng/ml) and RANKL (30 ng/ml) for another 6 days, to differentiate them into osteoclasts. Tartrate‐resistant alkaline phosphatase (TRAP)–positive multinucleated cells were quantified in the patient with Majeed syndrome, as well as in DIRA patients, healthy controls, and NOMID patients. B, Pit formation by osteoclasts was measured in macrophage cultures with fluoresceinamine‐labeled chondroitin sulfate/calcium phosphate. In A and B, results are based on 3 independent experiments, with values shown as the mean ± SD (healthy controls, n = 4; patient with Majeed syndrome, n = 1 with 2 visits; patients with NOMID, n = 3; and patients with DIRA, n = 3). C and D, M2‐MDMs were stained for RANK expression on day 7 of culture with M‐CSF (C) or stained for IL‐1 receptor (IL‐1R) (D). Nuclei were stained with DAPI. In C and D, results are based on 3 independent experiments, with values shown as the mean ± SD intensity, quantified using Imaris software (version 9.2.1) (healthy controls, n = 3; patient with Majeed syndrome, n = 1). Arrowheads show the altered surface distribution of IL‐1R in the patient’s M2‐MDMs compared to controls. In A–D, original magnification × 20. * = P < 0.05; ** = P < 0.001, by Student’s unpaired t‐test. NS = not significant (see Figure 2 for other definitions).
Figure 5
Figure 5
Effect of IL‐1 blockade on osteoclastogenesis and proinflammatory cytokine/chemokine production by M2‐MDMs. A, Monocyte‐derived M2‐MDMs were stimulated with LPS (1 μg/ml) and ATP (1 mmole) for 24 hours, and LPS‐ and ATP‐stimulated monocytes were cultured in the presence or absence of anakinra (10 μg/ml). The relative levels of various cytokines and chemokines in supernatant were analyzed by antibody array, and intensities of cytokine expression were analyzed using ImageJ software. Bars show the mean ± SD in cells from healthy controls (n = 2) (solid bars) and the patient with Majeed syndrome (n = 1 with 2 duplicates) (shaded bars). B, Monocytes from the patient with Majeed syndrome, patients with NOMID, and healthy controls were cultured with macrophage colony‐stimulating factor (M‐CSF) for 6 days, then RANKL was added to the M‐CSF, and, simultaneously, M2‐MDMs were cultured in the presence or absence of anakinra (1 mg/ml or 10 mg/ml). After 6 days, small and large tartrate‐resistant alkaline phosphatase–positive cells with >3 nuclei were quantified. Data are shown as box plots, where the boxes represent the 25th to 75th percentiles, the lines within the boxes represent the median, and the lines outside the boxes represent the minimum and maximum values (healthy controls [n = 3], NOMID patients [n = 3], patient with Majeed syndrome [n = 1 with 2 duplicates]). Original magnification × 20. * = P < 0.05; ** = P < 0.001. TNF = tumor necrosis factor; IP‐10 = interferon‐γ–inducible protein 10; GROα = growth‐related oncogene α; MCP‐1 = monocyte chemotactic protein 1; NS = not significant (see Figure 2 for other definitions).
Figure 6
Figure 6
IL‐1 and macrophage migration inhibitory factor (MIF) blockade inhibit osteoclastogenesis, and mutant lipin 2 regulates proinflammatory M2‐like macrophages (M2‐MDMs) by altering JNK/MAPK expression. A, IL‐1 inhibition with anakinra and MIF inhibition with ISO‐1 inhibited osteoclastogenesis most effectively. Macrophages were differentiated into osteoclasts. Cells were left untreated or treated with a MIF inhibitor (ISO‐1; 100 mM), an IL‐1 inhibitor (anakinra; 1 mg/ml), an IL‐6 inhibitor (tocilizumab; 10 mg/ml), a tumor necrosis factor inhibitor (SPD304; 5 mM), or a JNK inhibitor (SP600125; 10 mM). All experiments were performed in cells from healthy controls. Tartrate‐resistant alkaline phosphatase (TRAP)–positive multinucleated cells with >3 nuclei were quantified. Bars show the mean of triplicate assays. Original magnification × 100. B, The Human Phospho‐Kinase Array was used to detect multiple phosphorylated kinases in macrophages in the sample from the patient with Majeed syndrome (shaded bars) and healthy controls (solid bars), differentiated with macrophage colony‐stimulating factor (100 ng/ml) for 6 days. Cell lysate (60 μl) was analyzed to detect the phosphorylation state of M2‐MDMs at the basal level. Spot densities of phosphoproteins were quantified using ImageJ software. Bars show the mean ± SD of 2 independent experiments, with duplicates for the patient with Majeed syndrome. * = P < 0.05; ** = P < 0.001, by Student’s unpaired t‐test. GSK‐3 α/β = glycogen synthase kinase 3 α/β; TOR = mechanistic target of rapamycin (see Figure 2 for other definitions).

References

    1. De Jesus AA, Canna SW, Liu Y, Goldbach‐Mansky R. Molecular mechanisms in genetically defined autoinflammatory diseases: disorders of amplified danger signaling [review]. Annu Rev Immunol 2015;33:823–74.
    1. Ferguson PJ, Chen S, Tayeh MK, Ochoa L, Leal SM, Pelet A, et al. Homozygous mutations in LPIN2 are responsible for the syndrome of chronic recurrent multifocal osteomyelitis and congenital dyserythropoietic anaemia (Majeed syndrome). J Med Genet 2005;​42:551–7.
    1. Aksentijevich I, Masters SL, Ferguson PJ, Dancey P, Frenkel J, van Royen‐Kerkhoff A, et al. An autoinflammatory disease with deficiency of the interleukin‐1‐receptor antagonist. N Engl J Med 2009;360:2426–37.
    1. Reddy S, Jia S, Geoffrey R, Lorier R, Suchi M, Broeckel U, et al. An autoinflammatory disease due to homozygous deletion of the IL1RN locus. N Engl J Med 2009;360:2438–44.
    1. Majeed HA, Kalaawi M, Mohanty D, Teebi AS, Tunjekar MF, al‐Gharbawy F, et al. Congenital dyserythropoietic anemia and chronic recurrent multifocal osteomyelitis in three related children and the association with Sweet syndrome in two siblings. J Pediatr 1989;115:730–4.
    1. Herlin T, Fiirgaard B, Bjerre M, Kerndrup G, Hasle H, Bing X, et al. Efficacy of anti‐IL‐1 treatment in Majeed syndrome. Ann Rheum Dis 2013;72:410–3.
    1. Liu J, Hu XY, Zhao ZP, Guo RL, Guo J, Li W, et al. Compound heterozygous LPIN2 pathogenic variants in a patient with Majeed syndrome with recurrent fever and severe neutropenia: case report. BMC Med Genet 2019;20:182.
    1. Peterfy M, Phan J, Xu P, Reue K. Lipodystrophy in the fld mouse results from mutation of a new gene encoding a nuclear protein, lipin [letter]. Nat Genet 2001;27:121–4.
    1. Stein Y, Shapiro B. The synthesis of neutral glycerides by fractions of rat liver homogenates. Biochim Biophys Acta 1957;24:197–8.
    1. Han GS, Wu WI, Carman GM. The Saccharomyces cerevisiae Lipin homolog is a Mg2+‐dependent phosphatidate phosphatase enzyme. J Biol Chem 2006;281:9210–8.
    1. Kok BP, Venkatraman G, Capatos D, Brindley DN. Unlike two peas in a pod: lipid phosphate phosphatases and phosphatidate phosphatases [review]. Chem Rev 2012;112:5121–46.
    1. Reue K, Brindley DN. Thematic review series: glycerolipids. Multiple roles for lipins/phosphatidate phosphatase enzymes in lipid metabolism. J Lipid Res 2008;49:2493–503.
    1. Lorden G, Sanjuan‐Garcia I, de Pablo N, Meana C, Alvarez‐Miguel I, Perez‐Garcia MT, et al. Lipin‐2 regulates NLRP3 inflammasome by affecting P2X7 receptor activation. J Exp Med 2017;214:511–28.
    1. Hashimoto M, Nasser H, Chihara T, Suzu S. Macropinocytosis and TAK1 mediate anti‐inflammatory to pro‐inflammatory macrophage differentiation by HIV‐1 Nef. Cell Death Dis 2014;5:e1267.
    1. Verma SK, Chernomordik LV, Melikov K. An improved metrics for osteoclast multinucleation. Sci Rep 2018;8:1768.
    1. Hashimoto M, Nasser H, Bhuyan F, Kuse N, Satou Y, Harada S, et al. Fibrocytes differ from macrophages but can be infected with HIV‐1. J Immunol 2015;195:4341–50.
    1. Eckert MA, Coscia F, Chryplewicz A, Chang JW, Hernandez KM, Pan S, et al. Proteomics reveals NNMT as a master metabolic regulator of cancer‐associated fibroblasts. Nature 2019;569:723–8.
    1. Amarasekara DS, Yun H, Kim S, Lee N, Kim H, Rho J. Regulation of osteoclast differentiation by cytokine networks [review]. Immune Netw 2018;18:e8.
    1. Kwak HB, Ha H, Kim HN, Lee JH, Kim HS, Lee S, et al. Reciprocal cross‐talk between RANKL and interferon‐γ–inducible protein 10 is responsible for bone‐erosive experimental arthritis. Arthritis Rheum 2008;58:1332–42.
    1. Lee JH, Kim HN, Kim KO, Jin WJ, Lee S, Kim HH, et al. CXCL10 promotes osteolytic bone metastasis by enhancing cancer outgrowth and osteoclastogenesis. Cancer Res 2012;72:3175–86.
    1. Sucur A, Jajic Z, Artukovic M, Matijasevic MI, Anic B, Flegar D, et al. Chemokine signals are crucial for enhanced homing and differentiation of circulating osteoclast progenitor cells. Arthritis Res Ther 2017;19:142.
    1. Wiktor‐Jedrzejczak W, Bartocci A, Ferrante AW Jr, Ahmed‐Ansari A, Sell KW, Pollard JW, et al. Total absence of colony‐stimulating factor 1 in the macrophage‐deficient osteopetrotic (op/op) mouse. Proc Natl Acad Sci U S A 1990;87:4828–32.
    1. Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation. Nature 2003;423:337–42.
    1. Park JH, Lee NK, Lee SY. Current understanding of RANK signaling in osteoclast differentiation and maturation. Mol Cells 2017;40:706–13.
    1. Kim JH, Jin HM, Kim K, Song I, Youn BU, Matsuo K, et al. The mechanism of osteoclast differentiation induced by IL‐1. J Immunol 2009;183:1862–70.
    1. Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M, Yoshida H, et al. Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell 2002;3:889–901.
    1. Kim JH, Kim N. Regulation of NFATc1 in osteoclast differentiation. J Bone Metab 2014;21:233–41.
    1. Valdearcos M, Esquinas E, Meana C, Pena L, Gil‐de‐Gomez L, Balsinde J, et al. Lipin‐2 reduces proinflammatory signaling induced by saturated fatty acids in macrophages. J Biol Chem 2012;287:10894–904.
    1. David JP, Sabapathy K, Hoffmann O, Idarraga MH, Wagner EF. JNK1 modulates osteoclastogenesis through both c‐Jun phosphorylation‐dependent and ‐independent mechanisms. J Cell Sci 2002;115:4317–25.
    1. Ikeda F, Matsubara T, Tsurukai T, Hata K, Nishimura R, Yoneda T. JNK/c‐Jun signaling mediates an anti‐apoptotic effect of RANKL in osteoclasts. J Bone Miner Res 2008;23:907–14.
    1. Yamanaka Y, Abu‐Amer Y, Faccio R, Clohisy JC. Map kinase c‐JUN N‐terminal kinase mediates PMMA induction of osteoclasts. J Orthop Res 2006;24:1349–57.
    1. Li X, Udagawa N, Itoh K, Suda K, Murase Y, Nishihara T, et al. p38 MAPK‐mediated signals are required for inducing osteoclast differentiation but not for osteoclast function. Endocrinology 2002;143:3105–13.
    1. Kobayashi N, Kadono Y, Naito A, Matsumoto K, Yamamoto T, Tanaka S, et al. Segregation of TRAF6‐mediated signaling pathways clarifies its role in osteoclastogenesis. EMBO J 2001;20:1271–80.
    1. Teitelbaum SL, Ross FP. Genetic regulation of osteoclast development and function [review]. Nat Rev Genet 2003;4:638–49.
    1. Zhang Y, Tu Y, Zhao J, Chen K, Wu C. Reversion‐induced LIM interaction with Src reveals a novel Src inactivation cycle. J Cell Biol 2009;184:785–92.
    1. Bendre MS, Margulies AG, Walser B, Akel NS, Bhattacharrya S, Skinner RA, et al. Tumor‐derived interleukin‐8 stimulates osteolysis independent of the receptor activator of nuclear factor‐κB ligand pathway. Cancer Res 2005;65:11001–9.
    1. Li X, Loberg R, Liao J, Ying C, Snyder LA, Pienta KJ, et al. A destructive cascade mediated by CCL2 facilitates prostate cancer growth in bone. Cancer Res 2009;69:1685–92.
    1. Zhang J, Patel L, Pienta KJ. CC chemokine ligand 2 (CCL2) promotes prostate cancer tumorigenesis and metastasis [review]. Cytokine Growth Factor Rev 2010;21:41–8.
    1. Roodman GD. Biology of osteoclast activation in cancer. J Clin Oncol 2001;19:3562–71.
    1. Balboa MA, Balsinde J, Dennis EA. Involvement of phosphatidate phosphohydrolase in arachidonic acid mobilization in human amnionic WISH cells. J Biol Chem 1998;273:7684–90.
    1. Balboa MA, Balsinde J, Dennis EA, Insel PA. A phospholipase D‐mediated pathway for generating diacylglycerol in nuclei from Madin‐Darby canine kidney cells. J Biol Chem 1995;270:11738–40.
    1. Balsinde J, Balboa MA, Insel PA, Dennis EA. Differential regulation of phospholipase D and phospholipase A2 by protein kinase C in P388D1 macrophages. Biochem J 1997;321:805–9.
    1. Arthur JS. MSK activation and physiological roles. Front Biosci 2008;13:5866–79.
    1. Kitanaka T, Nakano R, Kitanaka N, Kimura T, Okabayashi K, Narita T, et al. JNK activation is essential for activation of MEK/ERK signaling in IL‐1β‐induced COX‐2 expression in synovial fibroblasts. Sci Rep 2017;7:39914.
    1. Hu X, Han C, Jin J, Qin K, Zhang H, Li T, et al. Integrin CD11b attenuates colitis by strengthening Src‐Akt pathway to polarize anti‐inflammatory IL‐10 expression. Sci Rep 2016;6:26252.
    1. Hu X, Wang H, Han C, Cao X. Src promotes anti‐inflammatory (M2) macrophage generation via the IL‐4/STAT6 pathway. Cytokine 2018;111:209–15.
    1. Buechler C, Ritter M, Orso E, Langmann T, Klucken J, Schmitz G. Regulation of scavenger receptor CD163 expression in human monocytes and macrophages by pro‐ and antiinflammatory stimuli. J Leukoc Biol 2000;67:97–103.
    1. Sanjay A, Houghton A, Neff L, DiDomenico E, Bardelay C, Antoine E, et al. Cbl associates with Pyk2 and Src to regulate Src kinase activity, α(v)β(3) integrin‐mediated signaling, cell adhesion, and osteoclast motility. J Cell Biol 2001;152:181–95.
    1. Zhao M, Finlay D, Zharkikh I, Vuori K. Novel role of Src in priming Pyk2 phosphorylation. PLoS One 2016;11:e0149231.
    1. Thomas SM, Brugge JS. Cellular functions regulated by Src family kinases. Annu Rev Cell Dev Biol 1997;13:513–609.
    1. Boyce BF, Yoneda T, Lowe C, Soriano P, Mundy GR. Requirement of pp60c‐src expression for osteoclasts to form ruffled borders and resorb bone in mice. J Clin Invest 1992;90:1622–7.
    1. Lowe C, Yoneda T, Boyce BF, Chen H, Mundy GR, Soriano P. Osteopetrosis in Src‐deficient mice is due to an autonomous defect of osteoclasts. Proc Natl Acad Sci U S A 1993;90:4485–9.
    1. Aleshin A, Finn RS. SRC: a century of science brought to the clinic [review]. Neoplasia 2010;12:599–607.
    1. Carron JD, Greinwald JH, Oberman JP, Werner AL, Derkay CS. Simulated reflux and laryngotracheal reconstruction: a rabbit model. Arch Otolaryngol Head Neck Surg 2001;127:576–80.
    1. Fregosi RF, Hwang JC, Bartlett D Jr, St. John WM. Activity of abdominal muscle motoneurons during hypercapnia. Respir Physiol 1992;89:179–94.
    1. Xiong WC, Feng X. PYK2 and FAK in osteoclasts. Front Biosci 2003;8:d1219–26.
    1. Miyazaki T, Sanjay A, Neff L, Tanaka S, Horne WC, Baron R. Src kinase activity is essential for osteoclast function. J Biol Chem 2004;​279:17660–6.
    1. Miyazaki T, Tanaka S, Sanjay A, Baron R. The role of c‐Src kinase in the regulation of osteoclast function. Mod Rheumatol 2006;​16:68–74.
    1. Ray BJ, Thomas K, Huang CS, Gutknecht MF, Botchwey EA, Bouton AH. Regulation of osteoclast structure and function by FAK family kinases. J Leukoc Biol 2012;92:1021–8.
    1. Nascimento EB, Snel M, Guigas B, van der Zon GC, Kriek J, Maassen JA, et al. Phosphorylation of PRAS40 on Thr246 by PKB/AKT facilitates efficient phosphorylation of Ser183 by mTORC1. Cell Signal 2010;22:961–7.
    1. Byles V, Covarrubias AJ, Ben‐Sahra I, Lamming DW, Sabatini DM, Manning BD, et al. The TSC‐mTOR pathway regulates macrophage polarization. Nat Commun 2013;4:2834.
    1. Kim JH, Sim JH, Lee S, Seol MA, Ye SK, Shin HM, et al. Interleukin‐7 induces osteoclast formation via STAT5, independent of receptor activator of NF‐κB ligand. Front Immunol 2017;8:1376.
    1. Granata F, Frattini A, Loffredo S, del Prete A, Sozzani S, Marone G, et al. Signaling events involved in cytokine and chemokine production induced by secretory phospholipase A2 in human lung macrophages. Eur J Immunol 2006;36:1938–50.
    1. Abe K, Cox A, Takamatsu N, Velez G, Laxer RM, Tse SM, et al. Gain‐of‐function mutations in a member of the Src family kinases cause autoinflammatory bone disease in mice and humans. Proc Natl Acad Sci U S A 2019;116:11872–7.
    1. Gu Q, Yang H, Shi Q. Macrophages and bone inflammation. J Orthop Translat 2017;10:86–93.
    1. Sinder BP, Pettit AR, McCauley LK. Macrophages: their emerging roles in bone [review]. J Bone Miner Res 2015;30:2140–9.
    1. Chang MK, Raggatt LJ, Alexander KA, Kuliwaba JS, Fazzalari NL, Schroder K, et al. Osteal tissue macrophages are intercalated throughout human and mouse bone lining tissues and regulate osteoblast function in vitro and in vivo. J Immunol 2008;181:​1232–44.

Source: PubMed

3
Prenumerera