Pivotal role of protein tyrosine phosphatase 1B (PTP1B) in the macrophage response to pro-inflammatory and anti-inflammatory challenge

P G Través, V Pardo, M Pimentel-Santillana, Á González-Rodríguez, M Mojena, D Rico, Y Montenegro, C Calés, P Martín-Sanz, A M Valverde, L Boscá, P G Través, V Pardo, M Pimentel-Santillana, Á González-Rodríguez, M Mojena, D Rico, Y Montenegro, C Calés, P Martín-Sanz, A M Valverde, L Boscá

Abstract

Inhibition of protein tyrosine phosphatase 1B (PTP1B) has been suggested as an attractive target to improve insulin sensitivity in different cell types. In the present work, we have investigated the effect of PTP1B deficiency on the response of human and murine macrophages. Using in vitro and in vivo approaches in mice and silencing PTP1B in human macrophages with specific siRNAs, we have demonstrated that PTP1B deficiency increases the effects of pro-inflammatory stimuli in both human and rodent macrophages at the time that decreases the response to alternative stimulation. Moreover, the absence of PTP1B induces a loss of viability in resting macrophages and mainly after activation through the classic pathway. Analysis of early gene expression in macrophages treated with pro-inflammatory stimuli confirmed this exacerbated inflammatory response in PTP1B-deficient macrophages. Microarray analysis in samples from wild-type and PTP1B-deficient macrophages obtained after 24 h of pro-inflammatory stimulation showed an activation of the p53 pathway, including the excision base repair pathway and the insulin signaling pathway in the absence of PTP1B. In animal models of lipopolysaccharide (LPS) and D-galactosamine challenge as a way to reveal in vivo inflammatory responses, animals lacking PTP1B exhibited a higher rate of death. Moreover, these animals showed an enhanced response to irradiation, in agreement with the data obtained in the microarray analysis. In summary, these results indicate that, although inhibition of PTP1B has potential benefits for the treatment of diabetes, it accentuates pro-inflammatory responses compromising at least macrophage viability.

Figures

Figure 1
Figure 1
Effect of PTP1B deficiency on the polarization of macrophages. Peritoneal macrophages from wild-type (WT) or PTP1B-deficient (KO) mice were isolated after thioglycollate eliciting. Cells were stimulated with 200 ng/ml of LPS or 25 μg/ml polyI:C (M1 stimuli) and the levels of the indicated proteins were determined at 24 h (a). The response to M2 stimuli was analyzed after challenge for 24 h with a mixture of 20 ng/ml each IL-4+IL-13 (b). The levels of PTP1B in M1 and M2 polarized macrophages were determined (c). The accumulation of NO, PGE2 and TNF-α in the incubation medium was determined after 24 h of activation with 200 ng/ml LPS, 25 μg/ml polyI:C, 5 μg/ml LTA, 20 ng/ml IL-10 or 20 ng/ml of IL-4+IL-13 (d). The time course of mRNA levels of IL-6 and NOS-2 after activation with 200 ng/ml LPS (e) and of cells undergoing apoptosis by annexin V binding criteria (f) were determined. Results show the mean±S.D. of three independent experiments. *P<0.05; **P<0.01 versus the same condition in the WT cells or non-treated cells (c). (ac) The band intensities of NOS-2, COX-2 (a), HO-1, Arg-1 (b) and PTP1B (c) after normalization using p85 and β-actin, respectively, are shown
Figure 2
Figure 2
PTP1B deficiency enhances IκBα degradation and impairs IκBα recovery. Macrophages from WT and PTP1B KO mice were stimulated with 200 ng/ml of LPS for the indicated times and the NF-κB and IRF3 pathways were analyzed (a). The upregulation of IκBα mRNA expression as a sensor of resetting of NF-κB was determined by qPCR (b). The nuclear translocation of p65 was determined by western blotting using p85 and lamin B as markers of the corresponding cytosolic and nuclear fractions, respectively (c). Results show the mean±S.D. of four independent experiments (b) or a representative blot out of four (a, c). *P<0.05; **P<0.01 versus the same condition in the WT cells
Figure 3
Figure 3
PTP1B deficiency enhances AKT and MAPK activation. Macrophages from WT and PTP1B KO mice were stimulated with 200 ng/ml of LPS and the phosphorylation of AKT, ERK, p38 and JNK was determined at the indicated times (a). To evaluate the effect of the PI3K pathway in the modulation of AKT and MAPK activities, macrophages from PTP1B KO mice were treated for 10 min before LPS activation with 10 μM LY29004. At the indicated times, the phosphorylation state of these proteins was determined (b). In parallel to the precedent assays, the levels of IκBα were determined using a lower LPS concentration (50 ng/ml) as stimulus (c). The effect of recombinant PTP1B on the dephosphorylation of extracts from WT and PTP1B macrophages activated for 30 min with 200 ng/ml of LPS was assayed in vitro after 30 min of incubation at 30 °C with recombinant PTP1B (d). Results show the mean±S.D. of three independent experiments, or a representative dephosphorylation experiment (d). *P<0.05; **P<0.01 versus the same condition in the WT cells
Figure 4
Figure 4
Gene expression profiling of WT and PTP1B-deficient macrophages treated with LPS. Macrophages were activated with 200 ng/ml of LPS for 24 h and the RNA was extracted, analyzed for quality in a Bioanalyzer and submitted to microarray hybridization (Agilent). Heatmap showing the microarray probes with differential response (FDRa). Gene set enrichment analysis (GSEA) of KEGG and REACTOME pathways in macrophages from PTP1B KO mice after pro-inflammatory stimulation (b). The genes in significantly enriched pathways (FDR<0.25) are shown and the color code corresponds to the standardized gene expression per row (not LPS fold change as in a). Red indicates higher expression and blue lower expression
Figure 5
Figure 5
Effect of PTP1B silencing on the response of human macrophages. Human monocytes from buffy coats were differentiated into macrophages and treated with a mixture of sc- or siRNA oligonucleotides to silence PTP1B. After incubation for 24 h with 200 ng/ml of LPS plus 20 ng/ml of human IFN-γ, 25 μg/ml of polyI:C or 20 ng/ml of each human IL-4+IL-13, the levels of PTP1B and COX2 were determined (a). The time course of the phosphorylation and total levels of proteins related to NF-κB, AKT and MAPKs were determined (b). The release of TNF-α and PGE2 to the culture medium was determined with specific kits (c). The induction of apoptosis, measured as the annexin V-positive population, was determined in both resting and LPS+IFN-γ activated cells (d). In addition to this, to determine the sensitivity of cells silenced for PTP1B to apoptosis in response to pro-inflammatory dependent stimuli the time course and the sensitivity at 36 h to different concentrations of staurosporine (as an stressor to promote mitochondrial-dependent apoptosis) were determined (d). Results show a representative experiment or the mean±S.D. of three independent experiments. *P<0.05; **P<0.01 versus the same condition in cells treated with scRNA
Figure 6
Figure 6
In vivo effect of PTP1B deficiency on the response of mice to different pro-inflammatory challenges. WT or PTP1B KO male mice matched in age were used in these experiments. The serum levels of TNF-α of untreated animals (n=12) were determined (a). The ear MPO activity and edema (n=10 animals per condition) after topic administration of TPA was calculated as ratio versus the ear treated with vehicle (b). Fixed ears were stained with eosin/hematoxylin to show the structure and with (sagittal sections; c). The serum levels of TNF-α of untreated animals or i.p. injected LPS (1 mg/kg body weight) or zymosan (30 mg/kg body weight) were determined at 1 h (n=7 animals per condition) (d). The sensitivity to irradiation after a unique dose of 10 Gy or two doses of 5 Gy administered in an interval of 4 h (n=15 per group) was evaluated as survival after 7 days post transplantation (Tx) of the corresponding bone marrow; P=0.0005 versus the WT at 10 Gy (e). The liver injury, determined by the measurement of the ALT activity in the serum (f) and structure (eosin/hematoxylin staining at 4 h; white bar=100 μm; g), and the survival of animals after i.p. administration of a single dose of LPS/D-GalN (n=12 animals per condition) were determined; P=0.074 versus the WT (h, left). Alternatively, a series of WT animals underwent irradiation (10 Gy) as depicted in (e), and were subsequently submitted to bone marrow transplantation from WT or PTP1B KO mice. After restitution of the immune system (12 weeks), these animals were submitted to LPS/D-GalN challenge and survival was determined (h, right). The TNF-α levels after 1 h of treatment with LPS/D-GalN were determined in serum from animals of (i). Results show the mean±S.D. of the indicated number of animals and/or the Kaplan–Meier representation of survival (e and h). *P<0.05; **P<0.01 versus the same condition in the WT animals or samples

References

    1. Haj FG, Markova B, Klaman LD, Bohmer FD, Neel BG. Regulation of receptor tyrosine kinase signaling by protein tyrosine phosphatase-1B. J Biol Chem. 2003;278:739–744.
    1. Salmeen A, Andersen JN, Myers MP, Tonks NK, Barford D. Molecular basis for the dephosphorylation of the activation segment of the insulin receptor by protein tyrosine phosphatase 1B. Mol Cell. 2000;6:1401–1412.
    1. Seely BL, Staubs PA, Reichart DR, Berhanu P, Milarski KL, Saltiel AR, et al. Protein tyrosine phosphatase 1B interacts with the activated insulin receptor. Diabetes. 1996;45:1379–1385.
    1. Elchebly M, Payette P, Michaliszyn E, Cromlish W, Collins S, Loy AL, et al. Increased insulin sensitivity and obesity resistance in mice lacking the protein tyrosine phosphatase-1B gene. Science. 1999;283:1544–1548.
    1. Haj FG, Zabolotny JM, Kim YB, Kahn BB, Neel BG. Liver-specific protein-tyrosine phosphatase 1B (PTP1B) re-expression alters glucose homeostasis of PTP1B-/-mice. J Biol Chem. 2005;280:15038–15046.
    1. Klaman LD, Boss O, Peroni OD, Kim JK, Martino JL, Zabolotny JM, et al. Increased energy expenditure, decreased adiposity, and tissue-specific insulin sensitivity in protein-tyrosine phosphatase 1B-deficient mice. Mol Cell Biol. 2000;20:5479–5489.
    1. Gonzalez-Rodriguez A, Mas-Gutierrez JA, Mirasierra M, Fernandez-Perez A, Lee YJ, KO HJ, et al. Essential role of protein tyrosine phosphatase 1B in obesity-induced inflammation and peripheral insulin resistance during aging. Aging Cell. 2012;11:284–296.
    1. Gonzalez-Rodriguez A, Escribano O, Alba J, Rondinone CM, Benito M, Valverde AM. Levels of protein tyrosine phosphatase 1B determine susceptibility to apoptosis in serum-deprived hepatocytes. J Cell Physiol. 2007;212:76–88.
    1. Adachi M, Sekiya M, Arimura Y, Takekawa M, Itoh F, Hinoda Y, et al. Protein-tyrosine phosphatase expression in pre-B cell NALM-6. Cancer Res. 1992;52:737–740.
    1. Yi T, Cleveland JL, Ihle JN. Identification of novel protein tyrosine phosphatases of hematopoietic cells by polymerase chain reaction amplification. Blood. 1991;78:2222–2228.
    1. Myers MP, Andersen JN, Cheng A, Tremblay ML, Horvath CM, Parisien JP, et al. TYK2 and JAK2 are substrates of protein-tyrosine phosphatase 1B. J Biol Chem. 2001;276:47771–47774.
    1. Aoki N, Matsuda T. A cytosolic protein-tyrosine phosphatase PTP1B specifically dephosphorylates and deactivates prolactin-activated STAT5a and STAT5b. J Biol Chem. 2000;275:39718–39726.
    1. Lu X, Malumbres R, Shields B, Jiang X, Sarosiek KA, Natkunam Y, et al. PTP1B is a negative regulator of interleukin 4-induced STAT6 signaling. Blood. 2008;112:4098–4108.
    1. Heinonen KM, Dube N, Bourdeau A, Lapp WS, Tremblay ML. Protein tyrosine phosphatase 1B negatively regulates macrophage development through CSF-1 signaling. Proc Natl Acad Sci USA. 2006;103:2776–2781.
    1. Eichhorst ST, Krueger A, Muerkoster S, Fas SC, Golks A, Gruetzner U, et al. Suramin inhibits death receptor-induced apoptosis in vitro and fulminant apoptotic liver damage in mice. Nat Med. 2004;10:602–609.
    1. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32:593–604.
    1. Mantovani A, Garlanda C, Locati M. Macrophage diversity and polarization in atherosclerosis: a question of balance. Arterioscler Thromb Vasc Biol. 2009;29:1419–1423.
    1. Xu H, An H, Hou J, Han C, Wang P, Yu Y, et al. Phosphatase PTP1B negatively regulates MyD88- and TRIF-dependent proinflammatory cytokine and type I interferon production in TLR-triggered macrophages. Mol Immunol. 2008;45:3545–3552.
    1. Laird MH, Rhee SH, Perkins DJ, Medvedev AE, Piao W, Fenton MJ, et al. TLR4/MyD88/PI3K interactions regulate TLR4 signaling. J Leukoc Biol. 2009;85:966–977.
    1. Diaz-Guerra MJ, Castrillo A, Martin-Sanz P, Bosca L. Negative regulation by phosphatidylinositol 3-kinase of inducible nitric oxide synthase expression in macrophages. J Immunol. 1999;162:6184–6190.
    1. Guha M, Mackman N. The phosphatidylinositol 3-kinase-Akt pathway limits lipopolysaccharide activation of signaling pathways and expression of inflammatory mediators in human monocytic cells. J Biol Chem. 2002;277:32124–32132.
    1. Fukao T, Koyasu S. PI3K and negative regulation of TLR signaling. Trends Immunol. 2003;24:358–363.
    1. Ruse M, Knaus UG. New players in TLR-mediated innate immunity: PI3K and small Rho GTPases. Immunol Res. 2006;34:33–48.
    1. Nieto-Vazquez I, Fernandez-Veledo S, de Alvaro C, Rondinone CM, Valverde AM, Lorenzo M. Protein-tyrosine phosphatase 1B-deficient myocytes show increased insulin sensitivity and protection against tumor necrosis factor-alpha-induced insulin resistance. Diabetes. 2007;56:404–413.
    1. Al-Lahham S, Roelofsen H, Rezaee F, Weening D, Hoek A, Vonk R, et al. Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur J Clin Invest. 2012;42:357–364.
    1. Al-Lahham SH, Roelofsen H, Priebe M, Weening D, Dijkstra M, Hoek A, et al. Regulation of adipokine production in human adipose tissue by propionic acid. Eur J Clin Invest. 2010;40:401–407.
    1. Gordon S, Pasare C, Medzhitov R. The macrophage: past, present and future. Toll-like receptors: linking innate and adaptive immunity. Eur J Immunol. 2007;37 (Suppl 1:S9–S17.
    1. Mantovani A. Molecular pathways linking inflammation and cancer. Curr Mol Med. 2010;10:369–373.
    1. Rhee I, Veillette A. Protein tyrosine phosphatases in lymphocyte activation and autoimmunity. Nat Immunol. 2012;13:439–447.
    1. Berdnikovs S, Pavlov VI, Abdala-Valencia H, McCary CA, Klumpp DJ, Tremblay ML, et al. PTP1B deficiency exacerbates inflammation and accelerates leukocyte trafficking in vivo. J Immunol. 2012;188:874–884.
    1. Carbone CJ, Zheng H, Bhattacharya S, Lewis JR, Reiter AM, Henthorn P, et al. Protein tyrosine phosphatase 1B is a key regulator of IFNAR1 endocytosis and a target for antiviral therapies. Proc Natl Acad Sci USA. 2012;109:19226–19231.
    1. Heinonen KM, Bourdeau A, Doody KM, Tremblay ML. Protein tyrosine phosphatases PTP-1B and TC-PTP play nonredundant roles in macrophage development and IFN-gamma signaling. Proc Natl Acad Sci USA. 2009;106:9368–9372.
    1. Zabolotny JM, Kim YB, Welsh LA, Kershaw EE, Neel BG, Kahn BB. Protein-tyrosine phosphatase 1B expression is induced by inflammation in vivo. J Biol Chem. 2008;283:14230–14241.
    1. Grant L, Shearer K, Czopek A, Lees E, Owen C, Agouni A, et al. Myeloid-cell protein tyrosine phosphatase-1B deficiency in mice protects against high-fat diet and lipopolysaccharide induced inflammation, hyperinsulinemia and endotoxemia through an IL10 STAT3 dependent mechanism. Diabetes. 2013.
    1. Zhang J, Wang B, Zhang W, Wei Y, Bian Z, Zhang CY, et al. Protein tyrosine phosphatase 1B deficiency ameliorates murine experimental colitis via the expansion of myeloid-derived suppressor cells. PLoS ONE. 2013;8:e70828.
    1. Gonzalez-Rodriguez A, Clampit JE, Escribano O, Benito M, Rondinone CM, Valverde AM. Developmental switch from prolonged insulin action to increased insulin sensitivity in protein tyrosine phosphatase 1B-deficient hepatocytes. Endocrinology. 2007;148:594–608.
    1. Rao KM. MAP kinase activation in macrophages. J Leukoc Biol. 2001;69:3–10.
    1. Traves PG, de Atauri P, Marin S, Pimentel-Santillana M, Rodriguez-Prados JC, Marín de Mas I, et al. Relevance of the MEK/ERK signaling pathway in the metabolism of activated macrophages: a metabolomic approach. J Immunol. 2012;188:1402–1410.
    1. Hazeki K, Nigorikawa K, Hazeki O. Role of phosphoinositide 3-kinase in innate immunity. Biol Pharm Bull. 2007;30:1617–1623.
    1. He R, Yu Z, He Y, Zeng LF, Xu J, Wu L, et al. Double click reaction for the acquisition of a highly potent and selective mPTPB inhibitor. ChemMedChem. 2010;5:2051–2056.
    1. Jiang ZX, Zhang ZY. Targeting PTPs with small molecule inhibitors in cancer treatment. Cancer Metastasis Rev. 2008;27:263–272.
    1. Liu S, Zhou B, Yang H, He Y, Jiang ZX, Kumar S, et al. Aryl vinyl sulfonates and sulfones as active site-directed and mechanism-based probes for protein tyrosine phosphatases. J Am Chem Soc. 2008;130:8251–8260.
    1. von Moltke J, Trinidad NJ, Moayeri M, Kintzer AF, Wang SB, van Rooijen N, et al. Rapid induction of inflammatory lipid mediators by the inflammasome in vivo. Nature. 2012;490:107–111.
    1. Rodriguez-Prados JC, Traves PG, Cuenca J, Rico D, Aragones J, Martín-Sanz P, et al. Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation. J Immunol. 2010;185:605–614.
    1. Diaz-Guerra MJ, Castrillo A, Martin-Sanz P, Bosca L. Negative regulation by protein tyrosine phosphatase of IFN-gamma-dependent expression of inducible nitric oxide synthase. J Immunol. 1999;162:6776–6783.
    1. Smyth GK.Limma: linear models for microarray dataIn: Bioinformatics and Computational Biology Solutions using R and Bioconductor Gentleman R, Carey V, Dudoit S, Irizarry R, Huber W, (eds)Springer: New York; 2005
    1. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003;34:267–273.
    1. Suzuki K, Ota H, Sasagawa S, Sakatani T, Fujikura T. Assay method for myeloperoxidase in human polymorphonuclear leukocytes. Anal Biochem. 1983;132:345–352.
    1. Gross S, Gammon ST, Moss BL, Rauch D, Harding J, Heinecke JW, et al. Bioluminescence imaging of myeloperoxidase activity in vivo. Nat Med. 2009;15:455–461.

Source: PubMed

3
Abonnieren