Transcriptomic signatures reveal immune dysregulation in human diabetic and idiopathic gastroparesis

Madhusudan Grover, Simon J Gibbons, Asha A Nair, Cheryl E Bernard, Adeel S Zubair, Seth T Eisenman, Laura A Wilson, Laura Miriel, Pankaj J Pasricha, Henry P Parkman, Irene Sarosiek, Richard W McCallum, Kenneth L Koch, Thomas L Abell, William J Snape, Braden Kuo, Robert J Shulman, Travis J McKenzie, Todd A Kellogg, Michael L Kendrick, James Tonascia, Frank A Hamilton, Gianrico Farrugia, NIDDK Gastroparesis Clinical Research Consortium (GpCRC), Jose Serrano, Linda Nguyen, William Hasler, Kristy Zodrow, Madhusudan Grover, Simon J Gibbons, Asha A Nair, Cheryl E Bernard, Adeel S Zubair, Seth T Eisenman, Laura A Wilson, Laura Miriel, Pankaj J Pasricha, Henry P Parkman, Irene Sarosiek, Richard W McCallum, Kenneth L Koch, Thomas L Abell, William J Snape, Braden Kuo, Robert J Shulman, Travis J McKenzie, Todd A Kellogg, Michael L Kendrick, James Tonascia, Frank A Hamilton, Gianrico Farrugia, NIDDK Gastroparesis Clinical Research Consortium (GpCRC), Jose Serrano, Linda Nguyen, William Hasler, Kristy Zodrow

Abstract

Background: Cellular changes described in human gastroparesis have revealed a role for immune dysregulation, however, a mechanistic understanding of human gastroparesis and the signaling pathways involved are still unclear.

Methods: Diabetic gastroparetics, diabetic non-gastroparetic controls, idiopathic gastroparetics and non-diabetic non-gastroparetic controls underwent full-thickness gastric body biopsies. Deep RNA sequencing was performed and pathway analysis of differentially expressed transcripts was done using Ingenuity®. A subset of differentially expressed genes in diabetic gastroparesis was validated in a separate cohort using QT-PCR.

Results: 111 genes were differentially expressed in diabetic gastroparesis and 181 in idiopathic gastroparesis with a log2fold difference of | ≥ 2| and false detection rate (FDR) < 5%. Top canonical pathways in diabetic gastroparesis included genes involved with macrophages, fibroblasts and endothelial cells in rheumatoid arthritis, osteoarthritis pathway and differential regulation of cytokine production in macrophages and T helper cells by IL-17A and IL-17F. Top canonical pathways in idiopathic gastroparesis included genes involved in granulocyte adhesion and diapedesis, agranulocyte adhesion and diapedesis, and role of macrophages, fibroblasts and endothelial cells in rheumatoid arthritis. Sixty-five differentially expressed genes (log2fold difference | ≥ 2|, FDR < 5%) were common in both diabetic and idiopathic gastroparesis with genes in the top 5 canonical pathways associated with immune signaling. 4/5 highly differentially expressed genes (SGK1, APOLD1, CXCR4, CXCL2, and FOS) in diabetic gastroparesis were validated in a separate cohort of patients using RT-PCR. Immune profile analysis revealed that genes associated with M1 (pro inflammatory) macrophages were enriched in tissues from idiopathic gastroparesis tissues compared to controls (p < 0.05).

Conclusions: Diabetic and idiopathic gastroparesis have both unique and overlapping transcriptomic signatures. Innate immune signaling likely plays a central role in pathogenesis of human gastroparesis.

Keywords: Diabetes mellitus; Macrophages; Next generation sequencing; RNA; Signaling.

Conflict of interest statement

Ethics approval and consent to participate

IRB approval obtained at all clinical sites and at Mayo Clinic as follows: Temple University Office for Human Subjects Protections (FWA00004964); California Pacific Medical Center Research Institute (FWA00000921); Texas Tech University Health Sciences Center (El Paso) (FWA 00006767); University of Mississippi Medical Center (FWA00003630) and Mayo Clinic (IRB 07–003371). Written informed consent was obtained for gastroparesis cases and verbal consent was obtained for controls as per approval by respective IRBs.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Heat maps of differentially expressed genes (Log2fold change | ≥ 2|, FDR < 0.05) in (a) Diabetic gastroparesis and (b) Idiopathic gastroparesis
Fig. 2
Fig. 2
a Venn diagram showing overlapping differentially expressed genes between diabetic gastroparetics and diabetic controls and idiopathic gastroparetics and idiopathic controls (Log2fold change | ≥ 2|, FDR < 0.05). b Heat map demonstrating these overlapping, differentially expressed genes between diabetic gastroparetics and diabetic controls and idiopathic gastroparetics and idiopathic controls (Log2fold change | ≥ 2|, FDR < 0.05). c Top canonical pathways linking the overlapping genes involved between the two comparisons. The horizontal bars represent total number of genes present in the pathway, scaled to 100%. The orange dots indicate the ratio of the overlapping differentially expressed genes that map to the pathway divided by the total number of genes present in the same pathway, e.g. > 25% for Granulocyte Adhesion and Diapedesis
Fig. 3
Fig. 3
RT-PCR validation of 5 genes differentially expressed in diabetic gastroparetics and diabetic controls by RNA seq in a different set of diabetic gastroparesis patients: Significant downregulation of APOLD1, apolipoprotein L domain containing 1; CXCR4, C-X-C motif chemokine receptor 4; CXCL2, C-X-C motif chemokine ligand 2; and FOS, Fos proto-oncogene in diabetic gastroparesis, as observed in the RNA seq analysis. SGK1, serum/glucocorticoid regulated kinase 1 expression was not statistically different on RT-PCR in the validation cohort, but had significantly lower log2fold changes in expression on RNA seq (1.77)
Fig. 4
Fig. 4
CIBERSORT analysis displaying distribution of patients with M1 and M2 macrophage associated genes in (a) Diabetic controls and diabetic gastroparesis: No differences were seen in % genes/subject associated with M1 or M2 macrophage phenotype (b) Idiopathic controls and idiopathic gastroparesis: Significantly greater number of idiopathic gastroparesis patients express % genes associated with an M1 (proinflammatory) macrophage phenotype (Mean (SD): 0.04 (0.03) % vs 0.004 (0.007) % M1 associated genes in idiopathic gastroparesis and idiopathic controls respectively, p = 0.02). No differences were seen in % genes/subject associated with M2 macrophage phenotype

References

    1. Wang YR, Fisher RS, Parkman HP. Gastroparesis-related hospitalizations in the United States: trends, characteristics, and outcomes, 1995-2004. Am J Gastroenterol. 2008;103(2):313–322. doi: 10.1111/j.1572-0241.2007.01658.x.
    1. Pasricha PJ, Camilleri M, Hasler WL, Parkman HP. White Paper AGA: Gastroparesis: clinical and regulatory insights for clinical trials. Clin Gastroenterol Hepatol. 2017;15(8):1184–1190. doi: 10.1016/j.cgh.2017.04.011.
    1. Grover M, Farrugia G, Lurken MS, Bernard CE, Faussone-Pellegrini MS, Smyrk TC, et al. Cellular changes in diabetic and idiopathic gastroparesis. Gastroenterology. 2011;140(5):1575–1585. doi: 10.1053/j.gastro.2011.01.046.
    1. Ordog T, Takayama I, Cheung WK, Ward SM, Sanders KM. Remodeling of networks of interstitial cells of Cajal in a murine model of diabetic gastroparesis. Diabetes. 2000;49(10):1731–1739. doi: 10.2337/diabetes.49.10.1731.
    1. Grover M, Bernard CE, Pasricha PJ, Lurken MS, Faussone-Pellegrini MS, Smyrk TC, et al. Clinical-histological associations in gastroparesis: results from the gastroparesis clinical research consortium. Neurogastroenterol Motil. 2012;24(6):531–539. doi: 10.1111/j.1365-2982.2012.01894.x.
    1. Faussone-Pellegrini MS, Grover M, Pasricha PJ, Bernard CE, Lurken MS, Smyrk TC, et al. Ultrastructural differences between diabetic and idiopathic gastroparesis. J Cell Mol Med. 2012;16(7):1573–1581. doi: 10.1111/j.1582-4934.2011.01451.x.
    1. Choi KM, Kashyap PC, Dutta N, Stoltz GJ, Ordog T, Shea Donohue T, et al. CD206-positive M2 macrophages that express heme oxygenase-1 protect against diabetic gastroparesis in mice. Gastroenterology. 2010;138(7):2399–2409. doi: 10.1053/j.gastro.2010.02.014.
    1. Cipriani G, Gibbons SJ, Verhulst PJ, Choi KM, Eisenman ST, Hein SS, et al. Diabetic Csf1(op/op) mice lacking macrophages are protected against the development of delayed gastric emptying. Cell Mol Gastroenterol Hepatol. 2016;2(1):40–47. doi: 10.1016/j.jcmgh.2015.09.001.
    1. Grover M, Bernard CE, Pasricha PJ, Parkman HP, Gibbons SJ, Tonascia J, et al. Diabetic and idiopathic gastroparesis is associated with loss of CD206-positive macrophages in the gastric antrum. Neurogastroenterol Motil. 2017;29(6). 10.1111/nmo.13018. Epub 2017 Jan 9.
    1. Kalari KR, Nair AA, Bhavsar JD, O'Brien DR, Davila JI, Bockol MA, et al. MAP-RSeq: Mayo analysis pipeline for RNA sequencing. BMC Bioinformatics. 2014;15:224. doi: 10.1186/1471-2105-15-224.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–140. doi: 10.1093/bioinformatics/btp616.
    1. Hart SN, Therneau TM, Zhang Y, Poland GA, Kocher JP. Calculating sample size estimates for RNA sequencing data. J Comput Biol. 2013;20(12):970–978. doi: 10.1089/cmb.2012.0283.
    1. Kramer A, Green J, Pollard J, Jr, Tugendreich S. Causal analysis approaches in ingenuity pathway analysis. Bioinformatics. 2014;30(4):523–530. doi: 10.1093/bioinformatics/btt703.
    1. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–457. doi: 10.1038/nmeth.3337.
    1. Cipriani G, Gibbons SJ, Kashyap PC, Farrugia G. Intrinsic gastrointestinal macrophages: their phenotype and role in gastrointestinal motility. Cell Mol Gastroenterol Hepatol. 2016;2(2):120–130. doi: 10.1016/j.jcmgh.2016.01.003.
    1. Eisenman ST, Gibbons SJ, Verhulst PJ, Cipriani G, Saur D, Farrugia G. Tumor necrosis factor alpha derived from classically activated "M1" macrophages reduces interstitial cell of Cajal numbers. Neurogastroenterol Motil. 2017;29(4). 10.1111/nmo.12984. Epub 2016 Oct 25.
    1. Bernard CE, Gibbons SJ, Mann IS, Froschauer L, Parkman HP, Harbison S, et al. Association of low numbers of CD206-positive cells with loss of ICC in the gastric body of patients with diabetic gastroparesis. Neurogastroenterol Motil. 2014;26(9):1275–1284. doi: 10.1111/nmo.12389.
    1. Hata K, Andoh A, Shimada M, Fujino S, Bamba S, Araki Y, et al. IL-17 stimulates inflammatory responses via NF-kappaB and MAP kinase pathways in human colonic myofibroblasts. Am J Physiol Gastrointest Liver Physiol. 2002;282(6):G1035–G1044. doi: 10.1152/ajpgi.00494.2001.
    1. Zhang Z, Andoh A, Inatomi O, Bamba S, Takayanagi A, Shimizu N, et al. Interleukin-17 and lipopolysaccharides synergistically induce cyclooxygenase-2 expression in human intestinal myofibroblasts. J Gastroenterol Hepatol. 2005;20(4):619–627. doi: 10.1111/j.1440-1746.2004.03748.x.
    1. Yagi Y, Andoh A, Inatomi O, Tsujikawa T, Fujiyama Y. Inflammatory responses induced by interleukin-17 family members in human colonic subepithelial myofibroblasts. J Gastroenterol. 2007;42(9):746–753. doi: 10.1007/s00535-007-2091-3.
    1. Miljkovic D, Cvetkovic I, Vuckovic O, Stosic-Grujicic S, Mostarica Stojkovic M, Trajkovic V. The role of interleukin-17 in inducible nitric oxide synthase-mediated nitric oxide production in endothelial cells. Cell Mol Life Sci. 2003;60(3):518–525. doi: 10.1007/s000180300043.
    1. Bain CC, Scott CL, Uronen-Hansson H, Gudjonsson S, Jansson O, Grip O, et al. Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal Immunol. 2013;6(3):498–510. doi: 10.1038/mi.2012.89.
    1. Auffray C, Sieweke MH, Geissmann F. Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol. 2009;27:669–692. doi: 10.1146/annurev.immunol.021908.132557.
    1. Martinez FO, Helming L, Milde R, Varin A, Melgert BN, Draijer C, et al. Genetic programs expressed in resting and IL-4 alternatively activated mouse and human macrophages: similarities and differences. Blood. 2013;121(9):e57–e69. doi: 10.1182/blood-2012-06-436212.
    1. Gross TJ, Kremens K, Powers LS, Brink B, Knutson T, Domann FE, et al. Epigenetic silencing of the human NOS2 gene: rethinking the role of nitric oxide in human macrophage inflammatory responses. J Immunol. 2014;192(5):2326–2338. doi: 10.4049/jimmunol.1301758.
    1. Raes G, Van den Bergh R, De Baetselier P, Ghassabeh GH, Scotton C, Locati M, et al. Arginase-1 and Ym1 are markers for murine, but not human, alternatively activated myeloid cells. J Immunol. 2005;174(11):6561. doi: 10.4049/jimmunol.174.11.6561.
    1. Mikkelsen HB. Interstitial cells of Cajal, macrophages and mast cells in the gut musculature: morphology, distribution, spatial and possible functional interactions. J Cell Mol Med. 2010;14(4):818–832. doi: 10.1111/j.1582-4934.2010.01025.x.
    1. Muller PA, Koscso B, Rajani GM, Stevanovic K, Berres ML, Hashimoto D, et al. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cell. 2014;158(2):300–313. doi: 10.1016/j.cell.2014.04.050.
    1. Coquenlorge S, Duchalais E, Chevalier J, Cossais F, Rolli-Derkinderen M, Neunlist M. Modulation of lipopolysaccharide-induced neuronal response by activation of the enteric nervous system. J Neuroinflammation. 2014;11:202. doi: 10.1186/s12974-014-0202-7.
    1. Green CL, Ho W, Sharkey KA, McKay DM. Dextran sodium sulfate-induced colitis reveals nicotinic modulation of ion transport via iNOS-derived NO. Am J Physiol Gastrointest Liver Physiol. 2004;287(3):G706–G714. doi: 10.1152/ajpgi.00076.2004.
    1. Hori M, Kita M, Torihashi S, Miyamoto S, Won KJ, Sato K, et al. Upregulation of iNOS by COX-2 in muscularis resident macrophage of rat intestine stimulated with LPS. Am J Physiol Gastrointest Liver Physiol. 2001;280(5):G930–G938. doi: 10.1152/ajpgi.2001.280.5.G930.
    1. Herring BP, Hoggatt AM, Gupta A, Griffith S, Nakeeb A, Choi JN et al. Idiopathic gastroparesis is associated with specific transcriptional changes in the gastric muscularis externa. Neurogastroenterol Motil. 2017; 2017 Oct 20. doi: 10.1111/nmo.13230. [Epub ahead of print].

Source: PubMed

3
Sottoscrivi