Induction of RET dependent and independent pro-inflammatory programs in human peripheral blood mononuclear cells from Hirschsprung patients

Marta Rusmini, Paola Griseri, Francesca Lantieri, Ivana Matera, Kelly L Hudspeth, Alessandra Roberto, Joanna Mikulak, Stefano Avanzini, Valentina Rossi, Girolamo Mattioli, Vincenzo Jasonni, Roberto Ravazzolo, William J Pavan, Alessio Pini-Prato, Isabella Ceccherini, Domenico Mavilio, Marta Rusmini, Paola Griseri, Francesca Lantieri, Ivana Matera, Kelly L Hudspeth, Alessandra Roberto, Joanna Mikulak, Stefano Avanzini, Valentina Rossi, Girolamo Mattioli, Vincenzo Jasonni, Roberto Ravazzolo, William J Pavan, Alessio Pini-Prato, Isabella Ceccherini, Domenico Mavilio

Abstract

Hirschsprung disease (HSCR) is a rare congenital anomaly characterized by the absence of enteric ganglia in the distal intestinal tract. While classified as a multigenic disorder, the altered function of the RET tyrosine kinase receptor is responsible for the majority of the pathogenesis of HSCR. Recent evidence demonstrate a strong association between RET and the homeostasis of immune system. Here, we utilize a unique cohort of fifty HSCR patients to fully characterize the expression of RET receptor on both innate (monocytes and Natural Killer lymphocytes) and adaptive (B and T lymphocytes) human peripheral blood mononuclear cells (PBMCs) and to explore the role of RET signaling in the immune system. We show that the increased expression of RET receptor on immune cell subsets from HSCR individuals correlates with the presence of loss-of-function RET mutations. Moreover, we demonstrate that the engagement of RET on PBMCs induces the modulation of several inflammatory genes. In particular, RET stimulation with glial-cell line derived neurotrophic factor family (GDNF) and glycosyl-phosphatidylinositol membrane anchored co-receptor α1 (GFRα1) trigger the up-modulation of genes encoding either for chemokines (CCL20, CCL2, CCL3, CCL4, CCL7, CXCL1) and cytokines (IL-1β, IL-6 and IL-8) and the down-regulation of chemokine/cytokine receptors (CCR2 and IL8-Rα). Although at different levels, the modulation of these "RET-dependent genes" occurs in both healthy donors and HSCR patients. We also describe another set of genes that, independently from RET stimulation, are differently regulated in healthy donors versus HSCR patients. Among these "RET-independent genes", there are CSF-1R, IL1-R1, IL1-R2 and TGFβ-1, whose levels of transcripts were lower in HSCR patients compared to healthy donors, thus suggesting aberrancies of inflammatory responses at mucosal level. Overall our results demonstrate that immune system actively participates in the physiopathology of HSCR disease by modulating inflammatory programs that are either dependent or independent from RET signaling.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Phenotypic distribution and levels of…
Figure 1. Phenotypic distribution and levels of expression of RET receptor on circulating immune cell subsets.
(A) Flow cytometric dot plot (upper line) and histogram (lower line) graphs showing a representative example from an healthy donor of CD14pos monocytes, CD56pos (NK cells), CD3pos (T cells) and CD20pos (B cells) lymphocytes expressing RET receptor (black line) compared to isotype control (gray shaded histograms). (B) Summary graph of dot plots with medians (horizontal black bars) showing the mean fluorescence intensities (MFIs) of RET receptor on immune cells (black symbols) compared to that of isotype controls (open white symbols) from 17 healthy donors.
Figure 2. Correlation between the RET receptor…
Figure 2. Correlation between the RET receptor expression and RET transcripts on four different cell lines.
(A) Flow cytometric histogram graphs showing the MFI levels of expression of RET receptor (black line) of 4 different cell lines. Gray shaded histograms represent the isotype controls. (B) Histogram bar graph showing the number of RET mRNA copies produced by the same cell lines displayed in panel A and analyzed within the same time-frame in culture. Values are normalized on SK-N-MC cell line of one experiment and are reported as fold increased in expression (2−ΔCt) as mean of three independent experiments. Of note, the level of RET receptor expressed on cell membrane significantly correlated with the amount of RET transcripts, as assessed by the Spearman rank test for correlation.
Figure 3. Expression of RET receptor on…
Figure 3. Expression of RET receptor on immune cells from HSCR patients associated with a single nucleotide polymorphism at exon 2.
Summary graph of statistical dot plots showing MFI values of RET receptor expressed on lymphocytes and monocytes from 50 HSCR patients with medians (horizontal black bars) either in the absence (A) or in the presence (B) of an association analysis stratification based on the genotype at the exon 2 of RET gene (SNP rs1800858). We did not detect any statistically significant association between the phenotypic distribution of RET receptor on immune cells with RET genotype at exon 2 SNP.
Figure 4. Expression of RET receptor on…
Figure 4. Expression of RET receptor on immune cells from HSCR patients associated with pathogenic mutations of RET gene.
Summary graph of statistical histogram bars (upper panel) and dot plots (lower panel) showing MFI values of RET expressed on monocytes, T, B and NK lymphocytes from 46 HSCR patients stratified on the basis of individuals either carrying (gray histogram bars in upper panel and black symbols in the lower panel) or not carrying (empty histogram bars in upper panel and empty symbols in the lower panel) pathogenic RET mutation. 4 HSCR patients were excluded from the analysis because they were carrying mutations of the RET gene with uncertain effects.
Figure 5. Modulation of RET-dependent genes.
Figure 5. Modulation of RET-dependent genes.
Colorimetric scale graph showing the fold increase (red) and decrease (green) of mRNA transcripts for those genes modulated in both PBMCs from healthy donors and HSCR patients following treatment with GDNF and GFRα1. The differences in fold changes of RET-dependent genes between the PBMCs of three healthy donors and three HSCR patients analyzed are indicated by means of 2−ΔΔCt values included in each square and by the ranges of color tonality of the same square.
Figure 6. Modulation of RET-independent genes.
Figure 6. Modulation of RET-independent genes.
Histogram bar graph showing the relative transcript levels of genes that, regardless of treatment with GDNF and GFRα1, are differently modulated in PBMCs from healthy donors and HSCR patients. The amounts of mRNA either in freshly purified (left part of the graph) or treated (right part of the graph) of PBMCs of healthy donors (white bars) and HSCR patients (black bars) were detected by Taqman Low Density Array (TLDA) card.

References

    1. Parisi MA, Kapur RP (2000) Genetics of Hirschsprung disease. Curr Opin Pediatr 12: 610–617.
    1. Badner JA, Sieber WK, Garver KL, Chakravarti A (1990) A genetic study of Hirschsprung disease. Am J Hum Genet 46: 568–580.
    1. Takahashi M, Ritz J, Cooper GM (1985) Activation of a novel human transforming gene, ret, by DNA rearrangement. Cell 42: 581–588.
    1. Pasini B, Hofstra RM, Yin L, Bocciardi R, Santamaria G, et al. (1995) The physical map of the human RET proto-oncogene. Oncogene 11: 1737–1743.
    1. Hofstra RM, Wu Y, Stulp RP, Elfferich P, Osinga J, et al. (2000) RET and GDNF gene scanning in Hirschsprung patients using two dual denaturing gel systems. Hum Mutat 15: 418–429.
    1. Attie T, Pelet A, Edery P, Eng C, Mulligan LM, et al. (1995) Diversity of RET proto-oncogene mutations in familial and sporadic Hirschsprung disease. Hum Mol Genet 4: 1381–1386.
    1. Angrist M, Bolk S, Thiel B, Puffenberger EG, Hofstra RM, et al. (1995) Mutation analysis of the RET receptor tyrosine kinase in Hirschsprung disease. Hum Mol Genet 4: 821–830.
    1. Pasini B, Borrello MG, Greco A, Bongarzone I, Luo Y, et al. (1995) Loss of function effect of RET mutations causing Hirschsprung disease. Nat Genet 10: 35–40.
    1. Carlomagno F, De Vita G, Berlingieri MT, de Franciscis V, Melillo RM, et al. (1996) Molecular heterogeneity of RET loss of function in Hirschsprung's disease. EMBO J 15: 2717–2725.
    1. Iwashita T, Asai N, Murakami H, Matsuyama M, Takahashi M (1996) Identification of tyrosine residues that are essential for transforming activity of the ret proto-oncogene with MEN2A or MEN2B mutation. Oncogene 12: 481–487.
    1. Amiel J, Sproat-Emison E, Garcia-Barcelo M, Lantieri F, Burzynski G, et al. (2008) Hirschsprung disease, associated syndromes and genetics: a review. J Med Genet 45: 1–14.
    1. Emison ES, McCallion AS, Kashuk CS, Bush RT, Grice E, et al. (2005) A common sex-dependent mutation in a RET enhancer underlies Hirschsprung disease risk. Nature 434: 857–863.
    1. Sancandi M, Griseri P, Pesce B, Patrone G, Puppo F, et al. (2003) Single nucleotide polymorphic alleles in the 5' region of the RET proto-oncogene define a risk haplotype in Hirschsprung's disease. J Med Genet 40: 714–718.
    1. Garcia-Barcelo M, Ganster RW, Lui VC, Leon TY, So MT, et al. (2005) TTF-1 and RET promoter SNPs: regulation of RET transcription in Hirschsprung's disease. Hum Mol Genet 14: 191–204.
    1. Fernandez RM, Boru G, Peciña A, Jones K, López-Alonso M, et al. (2005) Ancestral RET haplotype associated with Hirschsprung's disease shows linkage disequilibrium breakpoint at −1249. J Med Genet 42: 322–327.
    1. Emison ES, Garcia-Barcelo M, Grice EA, Lantieri F, Amiel J, et al. (2010) Differential contributions of rare and common, coding and noncoding Ret mutations to multifactorial Hirschsprung disease liability. Am J Hum Genet 87: 60–74.
    1. Griseri P, Bachetti T, Puppo F, Lantieri F, Ravazzolo R, et al. (2005) A common haplotype at the 5' end of the RET proto-oncogene, overrepresented in Hirschsprung patients, is associated with reduced gene expression. Hum Mutat 25: 189–195.
    1. Miao X, Leon TY, Ngan ES, So MT, Yuan ZW, et al. (2010) Reduced RET expression in gut tissue of individuals carrying risk alleles of Hirschsprung's disease. Hum Mol Genet 19: 1461–1467.
    1. Sribudiani Y, Metzger M, Osinga J, Rey A, Burns AJ, et al. (2011) Variants in RET associated with Hirschsprung's disease affect binding of transcription factors and gene expression. Gastroenterology 140: 572–582.e572.
    1. Leon TY, Ngan ES, Poon HC, So MT, Lui VC, et al. (2009) Transcriptional regulation of RET by Nkx2-1, Phox2b, Sox10, and Pax3. J Pediatr Surg 44: 1904–1912.
    1. Pachnis V, Mankoo B, Costantini F (1993) Expression of the c-ret proto-oncogene during mouse embryogenesis. Development 119: 1005–1017.
    1. Schuchardt A, D'Agati V, Larsson-Blomberg L, Costantini F, Pachnis V (1994) Defects in the kidney and enteric nervous system of mice lacking the tyrosine kinase receptor Ret. Nature 367: 380–383.
    1. de Groot JW, Links TP, Plukker JT, Lips CJ, Hofstra RM (2006) RET as a diagnostic and therapeutic target in sporadic and hereditary endocrine tumors. Endocr Rev 27: 535–560.
    1. Airaksinen MS, Titievsky A, Saarma M (1999) GDNF family neurotrophic factor signaling: four masters, one servant? Mol Cell Neurosci 13: 313–325.
    1. Manié S, Santoro M, Fusco A, Billaud M (2001) The RET receptor: function in development and dysfunction in congenital malformation. Trends Genet 17: 580–589.
    1. Le Douarin NM, Teillet MA (1973) The migration of neural crest cells to the wall of the digestive tract in avian embryo. J Embryol Exp Morphol 30: 31–48.
    1. Barembaum M, Bronner-Fraser M (2005) Early steps in neural crest specification. Semin Cell Dev Biol 16: 642–646.
    1. Knecht AK, Bronner-Fraser M (2002) Induction of the neural crest: a multigene process. Nat Rev Genet 3: 453–461.
    1. Yang C, Hutto D, Sah DW (2006) Distribution of GDNF family receptor alpha3 and RET in rat and human non-neural tissues. J Mol Histol 37: 69–77.
    1. Avantaggiato V, Dathan NA, Grieco M, Fabien N, Lazzaro D, et al. (1994) Developmental expression of the RET protooncogene. Cell Growth Differ 5: 305–311.
    1. Gattei V, Celetti A, Cerrato A, Degan M, De Iuliis A, et al. (1997) Expression of the RET receptor tyrosine kinase and GDNFR-alpha in normal and leukemic human hematopoietic cells and stromal cells of the bone marrow microenvironment. Blood 89: 2925–2937.
    1. Tsuzuki T, Takahashi M, Asai N, Iwashita T, Matsuyama M, et al. (1995) Spatial and temporal expression of the ret proto-oncogene product in embryonic, infant and adult rat tissues. Oncogene 10: 191–198.
    1. Nakayama S, Iida K, Tsuzuki T, Iwashita T, Murakami H, et al. (1999) Implication of expression of GDNF/Ret signalling components in differentiation of bone marrow haemopoietic cells. Br J Haematol 105: 50–57.
    1. Lindahl M, Poteryaev D, Yu L, Arumae U, Timmusk T, et al. (2001) Human glial cell line-derived neurotrophic factor receptor alpha 4 is the receptor for persephin and is predominantly expressed in normal and malignant thyroid medullary cells. J Biol Chem 276: 9344–9351.
    1. Veiga-Fernandes H, Coles MC, Foster KE, Patel A, Williams A, et al. (2007) Tyrosine kinase receptor RET is a key regulator of Peyer's patch organogenesis. Nature 446: 547–551.
    1. Vargas-Leal V, Bruno R, Derfuss T, Krumbholz M, Hohlfeld R, et al. (2005) Expression and function of glial cell line-derived neurotrophic factor family ligands and their receptors on human immune cells. J Immunol 175: 2301–2308.
    1. Borrello MG, Alberti L, Fischer A, Degl'innocenti D, Ferrario C, et al. (2005) Induction of a proinflammatory program in normal human thyrocytes by the RET/PTC1 oncogene. Proc Natl Acad Sci U S A 102: 14825–14830.
    1. Puppo F, Griseri P, Fanelli M, Schena F, Romeo G, et al. (2002) Cell-line specific chromatin acetylation at the Sox10-Pax3 enhancer site modulates the RET proto-oncogene expression. FEBS Lett 523: 123–127.
    1. Verbeek HH, Alves MM, de Groot JW, Osinga J, Plukker JT, et al. (2011) The effects of four different tyrosine kinase inhibitors on medullary and papillary thyroid cancer cells. J Clin Endocrinol Metab 96: E991–995.
    1. Lantieri F, Griseri P, Puppo F (2006) Campus R, Martucciello G, et al (2006) Haplotypes of the human RET proto-oncogene associated with Hirschsprung disease in the Italian population derive from a single ancestral combination of alleles. Ann Hum Genet 70: 12–26.
    1. Sunyaev S, Ramensky V, Koch I, Lathe W 3rd, Kondrashov AS, et al (2001) Prediction of deleterious human alleles. Hum Mol Genet 10: 591–597.
    1. Ng PC, Henikoff S (2003) SIFT: Predicting amino acid changes that affect protein function. Nucleic Acids Res 31: 3812–3814.
    1. Brunham LR, Singaraja RR, Pape TD, Kejariwal A, Thomas PD, et al. (2005) Accurate prediction of the functional significance of single nucleotide polymorphisms and mutations in the ABCA1 gene. PLoS Genet 1: e83.
    1. Thomas PD, Campbell MJ, Kejariwal A, Mi H, Karlak B, et al. (2003) PANTHER: a library of protein families and subfamilies indexed by function. Genome Res 13: 2129–2141.
    1. Cartegni L, Wang J, Zhu Z, Zhang MQ, Krainer AR (2003) ESEfinder: A web resource to identify exonic splicing enhancers. Nucleic Acids Res 31: 3568–3571.
    1. Reese MG, Eeckman FH, Kulp D, Haussler D (1997) Improved splice site detection in Genie. J Comput Biol 4: 311–323.
    1. Gabriel SB, Salomon R, Pelet A, Angrist M, Amiel J, et al. (2002) Segregation at three loci explains familial and population risk in Hirschsprung disease. Nat Genet 31: 89–93.
    1. Eketjall S, Ibanez CF (2002) Functional characterization of mutations in the GDNF gene of patients with Hirschsprung disease. Hum Mol Genet 11: 325–329.
    1. Pachnis V, Durbec P, Taraviras S, Grigoriou M, Natarajan D (1998) III. Role Of the RET signal transduction pathway in development of the mammalian enteric nervous system. Am J Physiol 275: G183–186.
    1. Costantini F, Shakya R (2006) GDNF/Ret signaling and the development of the kidney. Bioessays 28: 117–127.
    1. Kwon DH, Choi JY, Je YH, Lee SH (2012) The overexpression of acetylcholinesterase compensates for the reduced catalytic activity caused by resistance-conferring mutations in Tetranychus urticae. Insect Biochem Mol Biol 42: 212–219.
    1. Luzi E, Marini F, Tognarini I, Carbonell Sala S, Galli G, et al. (2010) Ribozyme-mediated compensatory induction of menin-oncosuppressor function in primary fibroblasts from MEN1 patients. Cancer Gene Ther 17: 814–825.
    1. Luciani A, Villella VR, Esposito S, Brunetti-Pierri N, Medina DL, et al. (2011) Cystic fibrosis: a disorder with defective autophagy. Autophagy 7: 104–106.
    1. Turner MD, Chaudhry A, Nedjai B (2012) Tumour necrosis factor receptor trafficking dysfunction opens the TRAPS door to pro-inflammatory cytokine secretion. Biosci Rep 32: 105–112.
    1. von Boyen GB, Schulte N, Pfluger C, Spaniol U, Hartmann C, et al. (2011) Distribution of enteric glia and GDNF during gut inflammation. BMC Gastroenterol 11: 3.
    1. Natarajan D, Marcos-Gutierrez C, Pachnis V, de Graaff E (2002) Requirement of signalling by receptor tyrosine kinase RET for the directed migration of enteric nervous system progenitor cells during mammalian embryogenesis. Development 129: 5151–5160.
    1. Cook DN, Prosser DM, Forster R, Zhang J, Kuklin NA, et al. (2000) CCR6 mediates dendritic cell localization, lymphocyte homeostasis, and immune responses in mucosal tissue. Immunity 12: 495–503.
    1. Varona R, Villares R, Carramolino L, Goya I, Zaballos A, et al. (2001) CCR6-deficient mice have impaired leukocyte homeostasis and altered contact hypersensitivity and delayed-type hypersensitivity responses. J Clin Invest 107: R37–45.
    1. Schutyser E, Struyf S, Van Damme J (2003) The CC chemokine CCL20 and its receptor CCR6. Cytokine Growth Factor Rev 14: 409–426.
    1. Lee HJ, Choi SC, Lee MH, Oh HM, Choi EY, et al. (2005) Increased expression of MIP-3alpha/CCL20 in peripheral blood mononuclear cells from patients with ulcerative colitis and its down-regulation by sulfasalazine and glucocorticoid treatment. Inflamm Bowel Dis 11: 1070–1079.
    1. Le Borgne M, Etchart N, Goubier A, Lira SA, Sirard JC, et al. (2006) Dendritic cells rapidly recruited into epithelial tissues via CCR6/CCL20 are responsible for CD8+ T cell crosspriming in vivo. Immunity 24: 191–201.
    1. Kaser A, Ludwiczek O, Holzmann S, Moschen AR, Weiss G, et al. (2004) Increased expression of CCL20 in human inflammatory bowel disease. J Clin Immunol 24: 74–85.
    1. Chuntharapai A, Kim KJ (1995) Regulation of the expression of IL-8 receptor A/B by IL-8: possible functions of each receptor. J Immunol 155: 2587–2594.
    1. Franci C, Gosling J, Tsou CL, Coughlin SR, Charo IF (1996) Phosphorylation by a G protein-coupled kinase inhibits signaling and promotes internalization of the monocyte chemoattractant protein-1 receptor. Critical role of carboxyl-tail serines/threonines in receptor function. J Immunol 157: 5606–5612.
    1. Volpe S, Cameroni E, Moepps B, Thelen S, Apuzzo T, et al. (2012) CCR2 Acts as Scavenger for CCL2 during Monocyte Chemotaxis. PLoS One 7: e37208.
    1. Zeilhofer HU, Schorr W (2000) Role of interleukin-8 in neutrophil signaling. Curr Opin Hematol 7: 178–182.
    1. Hume DA, MacDonald KP (2012) Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood 119: 1810–1820.
    1. O'Neill LA (2008) The interleukin-1 receptor/Toll-like receptor superfamily: 10 years of progress. Immunol Rev 226: 10–18.
    1. Padgett RW, Reiss M (2007) TGFbeta superfamily signaling: notes from the desert. Development 134: 3565–3569.
    1. Elhalaby EA, Teitelbaum DH, Coran AG, Heidelberger KP (1995) Enterocolitis associated with Hirschsprung's disease: a clinical histopathological correlative study. J Pediatr Surg 30: 1023–1026; discussion 1026–1027.
    1. Fujimoto T, Reen DJ, Puri P (1988) Inflammatory response in enterocolitis in the piebald lethal mouse model of Hirschsprung's disease. Pediatr Res 24: 152–155.
    1. Murphy F, Puri P (2005) New insights into the pathogenesis of Hirschsprung's associated enterocolitis. Pediatr Surg Int 21: 773–779.
    1. Dinarello CA (2009) Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 27: 519–550.
    1. Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, et al. (2005) Macrophage receptors and immune recognition. Annu Rev Immunol 23: 901–944.
    1. Li MO, Flavell RA (2008) TGF-beta: a master of all T cell trades. Cell 134: 392–404.
    1. Pini Prato A, Gentilino V, Giunta C, Avanzini S, Parodi S, et al. (2008) Hirschsprung's disease: 13 years' experience in 112 patients from a single institution. Pediatr Surg Int 24: 175–182.
    1. Brunetta E, Fogli M, Varchetta S, Bozzo L, Hudspeth KL, et al. (2009) The decreased expression of Siglec-7 represents an early marker of dysfunctional natural killer-cell subsets associated with high levels of HIV-1 viremia. Blood 114: 3822–3830.

Source: PubMed

3
Abonnieren