scholarly journals HIF-alpha Activation Impacts Macrophage Function During Murine Leishmania major Infection

Author(s):  
Manjunath Bettadapura ◽  
Hayden Roys ◽  
Anne Bowlin ◽  
Gopinath Venugopal ◽  
Charity L. Washam ◽  
...  

Leishmanial skin lesions are characterized by inflammatory hypoxia alongside the activation of hypoxia inducible factors, HIF-1a and HIF-2a, and subsequent expression of the HIF-a target VEGF-A during Leishmania major infection. However, the factors responsible for HIF-a activation are not known. We hypothesize hypoxia and pro-inflammatory stimuli contribute to HIF-a activation during infection. RNASeq on leishmanial lesions found transcripts associated with HIF-1a signaling are induced. To determine whether hypoxia contributes to HIF-a activation, we followed the fate of myeloid cells infiltrating from the blood and into hypoxic lesions. Recruited myeloid cells experience hypoxia when they enter inflamed lesions, and the length of time in lesions increases their hypoxic signature. To determine whether pro-inflammatory stimuli in the inflamed tissue can also influence HIF-a activation, we subjected macrophages to various pro-inflammatory stimuli and measured VEGF-A. While parasites alone did not induce VEGF-A, and pro-inflammatory stimuli only modestly induce VEGF-A, HIF- stabilization increases VEGF-A during infection. HIF-a stabilization does not impact parasite entry, growth or killing. Alternatively, the absence of ARNT/HIF- signaling enhances parasite internalization. Altogether, these findings suggest HIF-a is active during infection, and while macrophage HIF-a activation promotes lymphatic remodeling through VEGF-A production, HIF-a activation does not impact parasite internalization or control.

Pathogens ◽  
2021 ◽  
Vol 10 (12) ◽  
pp. 1584
Author(s):  
Manjunath Bettadapura ◽  
Hayden Roys ◽  
Anne Bowlin ◽  
Gopinath Venugopal ◽  
Charity L. Washam ◽  
...  

Leishmanial skin lesions are characterized by inflammatory hypoxia alongside the activation of hypoxia-inducible factors, HIF-1α and HIF-2α, and subsequent expression of the HIF-α target VEGF-A during Leishmania major infection. However, the factors responsible for HIF-α activation are not known. We hypothesize that hypoxia and proinflammatory stimuli contribute to HIF-α activation during infection. RNA-Seq of leishmanial lesions revealed that transcripts associated with HIF-1α signaling were induced. To determine whether hypoxia contributes to HIF-α activation, we followed the fate of myeloid cells infiltrating from the blood and into hypoxic lesions. Recruited myeloid cells experienced hypoxia when they entered inflamed lesions, and the length of time in lesions increased their hypoxic signature. To determine whether proinflammatory stimuli in the inflamed tissue can also influence HIF-α activation, we subjected macrophages to various proinflammatory stimuli and measured VEGF-A. While parasites alone did not induce VEGF-A, and proinflammatory stimuli only modestly induced VEGF-A, HIF-α stabilization increased VEGF-A during infection. HIF-α stabilization did not impact parasite entry, growth, or killing. Conversely, the absence of ARNT/HIF-α signaling enhanced parasite internalization. Altogether, these findings suggest that HIF-α is active during infection, and while macrophage HIF-α activation promotes lymphatic remodeling through VEGF-A production, HIF-α activation does not impact parasite internalization or control.


2007 ◽  
Vol 56 (3) ◽  
pp. 231-234 ◽  
Author(s):  
Yasuyuki Goto ◽  
Chizu Sanjoba ◽  
Nana Arakaki ◽  
Masaaki Okamoto ◽  
Keiichi Saeki ◽  
...  

2010 ◽  
Vol 83 (6) ◽  
pp. 1295-1300 ◽  
Author(s):  
Pieter-Paul van Thiel ◽  
Wendy F. van der Meide ◽  
Allard van der Sluis ◽  
Jimmy E. Zeegelaar ◽  
Aldert Bart ◽  
...  

2013 ◽  
Vol 95 (2) ◽  
pp. 347-355 ◽  
Author(s):  
Wânia F. Pereira-Manfro ◽  
Flávia L. Ribeiro-Gomes ◽  
Alessandra Almeida Filardy ◽  
Natália S. Vellozo ◽  
Landi V. C. Guillermo ◽  
...  

2011 ◽  
Vol 7 (9) ◽  
pp. 1220-1229 ◽  
Author(s):  
Guillermo Elizondo ◽  
Miriam Rodríguez-Sosa ◽  
Elizabet Estrada-Muñiz ◽  
Frank J. Gonzalez ◽  
Libia Vega

2009 ◽  
Vol 104 (6) ◽  
pp. 918-922 ◽  
Author(s):  
Silvia Dantas Cangussú ◽  
Carolina Carvalho de Souza ◽  
Camila França Campos ◽  
Leda Quercia Vieira ◽  
Luís Carlos Crocco Afonso ◽  
...  

2009 ◽  
Vol 55 (6) ◽  
pp. 705-713 ◽  
Author(s):  
Wenhui Wu ◽  
Luise Weigand ◽  
Susana Mendez

We have previously reported that vaccination with CpG oligodeoxynucleotides delivered concomitantly with live Leishmania major (Lm/CpG) eliminates lesions associated with live vaccination in C57BL/6 mice. The absence of lesions is at least in part a result of the CpG DNA-mediated activation of dermal dendritic cells to produce cytokines such as interleukin (IL)-6. Wild-type C57BL/6 mice and IL-6−/− mice were immunized with the Lm/CpG vaccine and monitored for the development of lesions. IL-6−/− mice developed extensive, nonhealing lesions following live vaccination. The analysis of the inoculation site and draining lymph nodes of the IL-6−/− mice revealed a constitutive reduction in lymphocyte numbers, particularly CD4+ T cells. Live vaccination resulted in the specific expansion of CD4+Foxp3+ regulatory T cells in the knockout mice, and in a decrease of CD4+ IFN-γ -producing cells. These results indicate that IL-6−/− mice may have collateral immune defects that could influence the development of the natural immune response to pathogens, vaccines, or other inflammatory stimuli.


Sign in / Sign up

Export Citation Format

Share Document