[HTML][HTML] B cells promote granulomatous inflammation during chronic Mycobacterium tuberculosis infection in mice

Y Chen, S Bharrhan, J Xu, T Sharma, Y Wang… - PLoS …, 2023 - journals.plos.org
Y Chen, S Bharrhan, J Xu, T Sharma, Y Wang, P Salgame, J Zhang, K Nargan, AJC Steyn
PLoS pathogens, 2023journals.plos.org
The current study reveals that in chronic TB, the B cell-deficient μMT strain, relative to wild-
type (WT) C57BL/6 mice, displays in the lungs lower levels of inflammation that are
associated with decreased CD4+ T cell proliferation, diminished Th1 response, and
enhanced levels of interleukin (IL)-10. The latter result raises the possibility that B cells may
restrict lung expression of IL-10 in chronic TB. These observations are recapitulated in WT
mice depleted for B cells using anti-CD20 antibodies. IL-10 receptor (IL-10R) blockade …
The current study reveals that in chronic TB, the B cell-deficient μMT strain, relative to wild-type (WT) C57BL/6 mice, displays in the lungs lower levels of inflammation that are associated with decreased CD4+ T cell proliferation, diminished Th1 response, and enhanced levels of interleukin (IL)-10. The latter result raises the possibility that B cells may restrict lung expression of IL-10 in chronic TB. These observations are recapitulated in WT mice depleted for B cells using anti-CD20 antibodies. IL-10 receptor (IL-10R) blockade reverses the phenotypes of decreased inflammation and attenuated CD4+ T cell responses in B cell-depleted mice. Together, these results suggest that in chronic murine TB, B cells, by virtue of their capacity to restrict expression of the anti-inflammatory and immunosuppressive IL-10 in the lungs, promote the development of a robust protective Th1 response, thereby optimizing anti-TB immunity. This vigorous Th1 immunity and restricted IL-10 expression may, however, allow the development of inflammation to a level that can be detrimental to the host. Indeed, decreased lung inflammation observed in chronically infected B cell-deficient mice, which exhibit augmented lung IL-10 levels, is associated with a survival advantage relative to WT animals. Collectively, the results reveal that in chronic murine TB, B cells play a role in modulating the protective Th1 immunity and the anti-inflammatory IL-10 response, which results in augmentation of lung inflammation that can be host-detrimental. Intriguingly, in tuberculous human lungs, conspicuous B cell aggregates are present in close proximity to tissue-damaging lesions manifesting necrosis and cavitation, suggesting the possibility that in human TB, B cells may contribute to the development of exacerbated pathology that is known to promote transmission. Since transmission is a major hindrance to TB control, investigating into whether B cells can shape the development of severe pulmonic pathological responses in tuberculous individuals is warranted.
PLOS