Innate immune responses to human rotavirus in the neonatal gnotobiotic piglet disease model

Immunology. 2010 Oct;131(2):242-56. doi: 10.1111/j.1365-2567.2010.03298.x.

Abstract

Intestinal and systemic dendritic cell (DC) frequencies, serum and small intestinal content cytokines and uptake/binding of human rotavirus (HRV) virus-like particles (VLP) were studied in HRV acutely infected or mock-inoculated neonatal gnotobiotic piglets. Intestinal, mesenteric lymph node (MLN) and splenic plasmacytoid DCs (pDCs), conventional DCs (cDCs) and macrophages/monocytes were assessed by flow cytometry. In infected pigs, serum and small intestinal content interferon-α (IFN-α) were highest, interleukin-12 (IL-12) was lower and IL-10, tumour necrosis factor-α and IL-6 were minimal. Compared with mock-inoculated piglets, frequencies of total intestinal DCs were higher; splenic and MLN DC frequencies were lower. Most intestinal pDCs, but few cDCs, were IFN-α(+) and intestinal macrophages/monocytes were negative for IFN-α. Serum IFN-α levels and IFN-α(+) intestinal pDCs were highly correlated, suggesting IFN-α production in vivo by intestinal pDCs (r=0·8; P<0·01). The intestinal pDCs and cDCs, but not intestinal macrophages/monocytes, of HRV-infected piglets showed significantly lower VLP uptake/binding compared with mock-inoculated piglets, suggesting higher activation of pDCs and cDCs in infected piglets. Both intestinal pDCs and cDCs were activated (IFN-α(+) and lower VLP binding) after HRV infection, suggesting their role in induction of HRV-specific immunity. Dose-effects of HRV on serum IFN-α and IFN-α(+) DCs were studied by infecting piglets with 100-fold higher HRV dose. A high dose increased parameters associated with inflammation (diarrhoea, intestinal pathology) but serum IFN-α and IFN-α(+) DCs were similar between both groups. The pDCs have both anti- and pro-inflammatory functions. Stimulation of the anti-inflammatory effects of pDCs after the high dose, without increasing their pro-inflammatory impacts, may be critical to reduce further immunopathology during HRV infection.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Animals, Newborn
  • Cell Count
  • Cytokines / blood
  • Dendritic Cells / cytology
  • Dendritic Cells / immunology
  • Dendritic Cells / metabolism
  • Dendritic Cells / virology
  • Diarrhea / diagnosis
  • Diarrhea / etiology
  • Disease Models, Animal
  • Feces / virology
  • Germ-Free Life / immunology*
  • Humans
  • Immunity, Innate / immunology*
  • Interferon-alpha / blood
  • Interferon-alpha / metabolism
  • Intestines / cytology
  • Intestines / immunology
  • Intestines / pathology
  • Jejunum / pathology
  • Lymph Nodes / cytology
  • Lymph Nodes / immunology
  • Macrophages / cytology
  • Macrophages / immunology
  • Macrophages / metabolism
  • Monocytes / cytology
  • Monocytes / immunology
  • Monocytes / metabolism
  • Rotavirus / genetics
  • Rotavirus / immunology
  • Rotavirus Infections / blood
  • Rotavirus Infections / complications
  • Rotavirus Infections / immunology*
  • Spleen / cytology
  • Spleen / immunology
  • Sus scrofa
  • Vaccines, Virus-Like Particle / genetics
  • Vaccines, Virus-Like Particle / metabolism
  • Viral Nonstructural Proteins / genetics
  • Virus Replication / genetics
  • Virus Shedding

Substances

  • Cytokines
  • Interferon-alpha
  • NSP3 protein, Rotavirus
  • Vaccines, Virus-Like Particle
  • Viral Nonstructural Proteins