scholarly journals Viral myocarditis involves the generation of autoreactive T cells with multiple antigen specificities that localize in lymphoid and non-lymphoid organs in the mouse model of CVB3 infection

2020 ◽  
Vol 124 ◽  
pp. 218-228
Author(s):  
Rakesh H. Basavalingappa ◽  
Rajkumar Arumugam ◽  
Ninaad Lasrado ◽  
Bharathi Yalaka ◽  
Chandirasegaran Massilamany ◽  
...  
Diabetologia ◽  
2014 ◽  
Vol 58 (1) ◽  
pp. 140-148 ◽  
Author(s):  
Yuxing Zhao ◽  
Nicholas A. Scott ◽  
Stacey Fynch ◽  
Lorraine Elkerbout ◽  
W. Wei-Lynn Wong ◽  
...  

2019 ◽  
Vol 234 (7) ◽  
pp. 11986-11998 ◽  
Author(s):  
Davood Rostamzadeh ◽  
Mohammad Reza Haghshenas ◽  
Farhad Daryanoosh ◽  
Mahdi Samadi ◽  
Ahmad Hosseini ◽  
...  

Viruses ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 1222
Author(s):  
Sandra Pinkert ◽  
Meike Kespohl ◽  
Nicolas Kelm ◽  
Ziya Kaya ◽  
Arnd Heuser ◽  
...  

Infection of mice with Coxsackievirus B3 (CVB3) triggers inflammation of the heart and this mouse model is commonly used to investigate underlying mechanisms and therapeutic aspects for viral myocarditis. Virus-triggered cytotoxicity and the activity of infiltrating immune cells contribute to cardiac tissue injury. In addition to cardiac manifestation, CVB3 causes cell death and inflammation in the pancreas. The resulting pancreatitis represents a severe burden and under such experimental conditions, analgesics may be supportive to improve the animals’ well-being. Notably, several known mechanisms exist by which analgesics can interfere with the immune system and thereby compromise the feasibility of the model. We set up a study aiming to improve animal welfare while ensuring model integrity and investigated how tramadol, an opioid, affects virus-induced pathogenicity and immune response in the heart. Tramadol was administered seven days prior to a CVB3 infection in C57BL/6 mice and treatment was continued until the day of analysis. Tramadol had no effect on the virus titer or viral pathogenicity in the heart tissue and the inflammatory response, a hallmark of myocardial injury, was maintained. Our results show that tramadol exerts no disruptive effects on the CVB3 myocarditis mouse model and, therefore, the demonstrated protocol should be considered as a general analgesic strategy for CVB3 infection.


2021 ◽  
Vol 27 (Supplement_1) ◽  
pp. S33-S34
Author(s):  
Rafael Czepielewski ◽  
Emma Erlich ◽  
Emily Onufer ◽  
Shannon Young ◽  
Ki-Wook Kim ◽  
...  

Abstract Pioneer reports of Crohn’s disease (CD) suggested that impaired lymphatic flow might drive its pathogenesis but remains unsettled. Nodules of tertiary lymphoid organs (TLO) are found in association with collecting lymphatic vessels (CLVs) of the mesentery that normally conducts lymph outflow from the intestine. Whether TLOs affect lymph transport is unknown. In the TNFΔARE mouse model of Crohn’s-like ileitis, TLOs are found in valves regions. Using lymphatic reporters and photoconversion to study cell trafficking from the intestine, our findings indicated that TLOs halts immune cells traveling from the inflamed ileum to the lymph node, effectively trapping DCs, B, and T cells, and impacting the development of microbe tolerogenic regulatory T cells. Lymphatic transport defects were intrinsic to the CLVs because the soluble fluorescent tracer’s passage through TLO was also blocked. Lymph blockage promoted retrograde lymphatic flow returning towards the gut wall due to incapable valves. Moreover, significant lymph leakage was found, specifically at the TLOs. Neutralizing anti-TNF mAb treatment into TNFΔARE mice is ineffective in eliminating TLOs or restoring lymphatic trafficking when administered in female mice with advanced disease. In males, the therapy was able to restore forward flow to the lymph node. However, even in the presence of TNF inhibition, both sexes demonstrated TLO lymph leakage. Thus, mesenteric TLOs that form during chronic ileitis drive broadly impaired lymph transit of molecules and cells from the intestine that is only partially reversible by neutralizing the cytokine cascade underlying the disease and establish a perennial tissue alteration.


2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Troy Wesson ◽  
Adeeba Dhalech ◽  
Christopher M. Robinson

Background and Hypothesis:Coxsackievirus B3 (CVB3) is a non-enveloped RNA virus from the Picornaviridae family and is a primary cause of viral myocarditis in the United States. Approximately 5% of all symptomatic CVB3 infections are fatal. Therefore, there is a need to identify the mechanism(s) that regulate a protective immune response to CVB3. However, viral epitopes that stimulate T cell responses to CVB3 remain poorly characterized. To this end, we used a mouse model of CVB3 infection to identify the viral immunogenic CD8 T cell epitopes. We hypothesized that isolated antigen-experienced CD8 T cells from infected mice would be stimulated in the presence of predicted viral epitopes, confirming CVB3-specific T cells. Experimental Design: To identify novel CD8 T cell epitopes, predicted 9-mer MHC binding peptides from the CVB3-Nancy polyprotein were identified using the Immune Epitope Database (IEDB) analysis resource consensus tool. The top ten predicted peptides were synthesized for our assays. Splenocytes from CVB3-infected male and female IFNAR -/- mice were stimulated with each peptide in the presence of brefeldin A for 6 hours at 37˚C. Following stimulation, cells were surfaced stained with antibodies specific for antigen-experienced CD8 T cells. Next, we performed intracellular staining for IFN-gamma. Cells were analyzed using flow cytometry. Candidate epitopes were identified as having results ≥2 standard deviations over the control. Results: Thus far, our analysis has revealed responses to three novel CD8 T cell epitopes within the peptide library, including the viral epitopes within VP1 protein and the RNA-dependent RNA polymerase. Conclusion and Potential ImpactOverall, these data provide an advancement in CVB3 immunology. Further, these data generate new tools like MHC-tetramers to track endogenous T cell responses to CVB3 infection.


2002 ◽  
Vol 41 (03) ◽  
pp. 129-134 ◽  
Author(s):  
A. Wolski ◽  
E. Palombo-Kinne ◽  
F. Wolf ◽  
F. Emmrich ◽  
W. Becker ◽  
...  

Summary Aim: The cellular joint infiltrate in rheumatoid arthritis patients is rich in CD4-positive T-helper lymphocytes and macrophages, rendering anti-CD4 monoclonal antibodies (mAbs) suitable for specific immunoscintigraphy of human/ experimental arthritis. Following intravenous injection, however, mAbs are present both in the free form and bound to CD4-positive, circulating monocytes and T-cells. Thus, the present study aimed at analyzing the relative contribution of the free and the cell-bound component to the imaging of inflamed joints in experimental adjuvant arthritis (AA). Methods: AA rat peritoneal macrophages or lymph node T-cells were incubated in vitro with saturating amounts of 99mTc-anti-CD4 mAb (W3/25) and injected i.v. into rats with AA. Results: In vitro release of 99mTc-anti-CD4 mAb from the cells was limited (on average 1.57%/h for macrophages and 0.84%/h for T-cells). Following i.v. injection, whole body/joint scans and tissue measurements showed only negligible accumulation of radioactivity in inflamed ankle joints (tissue: 0.22 and 0.34% of the injected activity, respectively), whereas the radioactivity was concentrated in liver (tissue: 79% and 71%, respectively), kidney, and urinary bladder. Unlike macrophages, however, anti-CD4 mAb-coated T-cells significantly accumulated in lymphoid organs, the inflamed synovial membrane of the ankle joints, as well as in elbow and knee joints. Conclusion: While the overall contribution of cell-bound mAbs to the imaging of arthritic joints with anti-CD4 mAbs is minimal, differential accumulation of macrophages and T-cells in lymphoid organs and the inflamed synovial membrane indicates preferential migration patterns of these 2 cell populations in arthritic rats. Although only validated for 99mTc-anti-CD4 mAbs, extrapolation of the results to other anticellular mAbs with similar affinity for their antigen may be possible.


Sign in / Sign up

Export Citation Format

Share Document