Use my Search Websuite to scan PubMed, PMCentral, Journal Hosts and Journal Archives, FullText.
Kick-your-searchterm to multiple Engines kick-your-query now !>
A dictionary by aggregated review articles of nephrology, medicine and the life sciences
Your one-stop-run pathway from word to the immediate pdf of peer-reviewed on-topic knowledge.

suck abstract from ncbi


10.3390/v13122383

http://scihub22266oqcxt.onion/10.3390/v13122383
suck pdf from google scholar
34960652!8708337!34960652
unlimited free pdf from europmc34960652    free
PDF from PMC    free
html from PMC    free

suck abstract from ncbi


Deprecated: Implicit conversion from float 217.6 to int loses precision in C:\Inetpub\vhosts\kidney.de\httpdocs\pget.php on line 534

Deprecated: Implicit conversion from float 217.6 to int loses precision in C:\Inetpub\vhosts\kidney.de\httpdocs\pget.php on line 534
pmid34960652      Viruses 2021 ; 13 (12): ä
Nephropedia Template TP

gab.com Text

Twit Text FOAVip

Twit Text #

English Wikipedia


  • Inhibiting ACSL1-Related Ferroptosis Restrains Murine Coronavirus Infection #MMPMID34960652
  • Xia H; Zhang Z; You F
  • Viruses 2021[Nov]; 13 (12): ä PMID34960652show ga
  • Murine hepatitis virus strain A59 (MHV-A59) was shown to induce pyroptosis, apoptosis, and necroptosis of infected cells, especially in the murine macrophages. However, whether ferroptosis, a recently identified form of lytic cell death, was involved in the pathogenicity of MHV-A59 is unknown. We utilized murine macrophages and a C57BL/6 mice intranasal infection model to address this. In primary macrophages, the ferroptosis inhibitor inhibited viral propagation, inflammatory cytokines released, and cell syncytia formed after MHV-A59 infection. In the mouse model, we found that in vivo administration of liproxstatin-1 ameliorated lung inflammation and tissue injuries caused by MHV-A59 infection. To find how MHV-A59 infection influenced the expression of ferroptosis-related genes, we performed RNA-seq in primary macrophages and found that MHV-A59 infection upregulates the expression of the acyl-CoA synthetase long-chain family member 1 (ACSL1), a novel ferroptosis inducer. Using ferroptosis inhibitors and a TLR4 inhibitor, we showed that MHV-A59 resulted in the NF-kB-dependent, TLR4-independent ACSL1 upregulation. Accordingly, ACSL1 inhibitor Triacsin C suppressed MHV-A59-infection-induced syncytia formation and viral propagation in primary macrophages. Collectively, our study indicates that ferroptosis inhibition protects hosts from MHV-A59 infection. Targeting ferroptosis may serve as a potential treatment approach for dealing with hyper-inflammation induced by coronavirus infection.
  • |*Ferroptosis[MESH]
  • |Animals[MESH]
  • |Coenzyme A Ligases/*antagonists & inhibitors/genetics/*metabolism[MESH]
  • |Coronavirus Infections/*therapy[MESH]
  • |Cytokines/metabolism[MESH]
  • |Disease Models, Animal[MESH]
  • |Genes, Viral[MESH]
  • |Lung Injury/pathology[MESH]
  • |Macrophages[MESH]
  • |Mice[MESH]
  • |Mice, Inbred C57BL[MESH]
  • |Murine hepatitis virus[MESH]
  • |Quinoxalines[MESH]
  • |RAW 264.7 Cells[MESH]
  • |Spiro Compounds[MESH]
  • |Toll-Like Receptor 4[MESH]


  • DeepDyve
  • Pubget Overpricing
  • suck abstract from ncbi

    Linkout box