Autoantigen-specific immune tolerance in pathological and physiological cell death: Nanotechnology comes into view

2020 ◽  
pp. 107177
Author(s):  
Amir Tajbakhsh ◽  
Najmeh Farahani ◽  
Sayed Mohammad Gheibihayat ◽  
Amir Masoud Mirkhabbaz ◽  
Amir Savardashtaki ◽  
...  
1991 ◽  
pp. 451-459
Author(s):  
R. A. Lockshin ◽  
Z. F. Zakeri ◽  
L. M. Yesner

2007 ◽  
Vol 236 (3) ◽  
pp. 880-885 ◽  
Author(s):  
V. Zuzarte-Luis ◽  
J.A. Montero ◽  
N. Torre-Perez ◽  
J.A. Garcia-Porrero ◽  
J.M. Hurle

1996 ◽  
Vol 4 (4-5) ◽  
pp. 341-353 ◽  
Author(s):  
Dorit Hanein ◽  
Anat Yarden ◽  
Helena Sabanay ◽  
Lia Addadi ◽  
Benjamin Geiger

1995 ◽  
Vol 15 (4) ◽  
pp. 299-311 ◽  
Author(s):  
Georg Häucker ◽  
David L. Vaux

2006 ◽  
Vol 926 (1) ◽  
pp. 83-89 ◽  
Author(s):  
PHILIPPE BOUILLET ◽  
DAVID C.S. HUANG ◽  
LORRAINE A. O'REILLY ◽  
HAMSA PUTHALAKATH ◽  
LIAM O'CONNOR ◽  
...  

1992 ◽  
Vol 12 (7) ◽  
pp. 3060-3069 ◽  
Author(s):  
D S Ucker ◽  
P S Obermiller ◽  
W Eckhart ◽  
J R Apgar ◽  
N A Berger ◽  
...  

We examined virally transformed murine fibroblast clones as targets for cytotoxic T lymphocyte (CTL)-triggered lysis and genome digestion. Strikingly, while all clones were essentially equivalent in the ability to be lysed, one clone, SV3T3-B2.1, failed to exhibit genome digestion associated with CTL attack. Other aspects of the physiological cell death process, including loss of adhesion and nuclear envelope breakdown (lamin phosphorylation and solubilization), were not altered in this clone. The absence of genome digestion associated with CTL-induced cell death correlated with the absence of endodeoxyribonuclease activity in the nuclei of that clone. Characterization of the activity affected identifies a calcium-dependent, DNase I-like endonuclease of approximately 40 kDa, normally present constitutively in all cell nuclei, as the enzyme responsible for genome digestion associated with CTL-mediated cell death. These observations indicate that neither genome digestion per se nor its consequences [such as activation of poly(ADP-ribose) polymerase] are essential for cell death resulting from the triggering of this cell suicide process.


2019 ◽  
Vol 2019 ◽  
pp. 1-11 ◽  
Author(s):  
Ana Carolina Martínez-Torres ◽  
Kenny Misael Calvillo-Rodríguez ◽  
Ashanti Concepción Uscanga-Palomeque ◽  
Luis Gómez-Morales ◽  
Rodolfo Mendoza-Reveles ◽  
...  

Acute lymphocytic leukemia (ALL) is the most common pediatric cancer. Currently, treatment options for patients with relapsed and refractory ALL mostly rely on immunotherapies. However, hematological cancers are commonly associated with a low immunogenicity and immune tolerance, which may contribute to leukemia relapse and the difficulties associated with the development of effective immunotherapies against this disease. We recently demonstrated that PKHB1, a TSP1-derived CD47 agonist peptide, induces immunogenic cell death (ICD) in T cell ALL (T-ALL). Cell death induced by PKHB1 on T-ALL cell lines and their homologous murine, L5178Y-R (T-murine tumor lymphoblast cell line), induced damage-associated molecular patterns (DAMPs) exposure and release. Additionally, a prophylactic vaccination with PKHB1-treated L5178Y-R cells prevented tumor establishment in vivo in all the cases. Due to the immunogenic potential of PKHB1-treated cells, in this study we assessed their ability to induce antitumor immune responses ex vivo and in vivo in an established tumor. We first confirmed the selectivity of cell death induced by PKBH1 in tumor L5178Y-R cells and observed that calreticulin exposure increased when cell death increased. Then, we found that the tumor cell lysate (TCL) obtained from PKHB1-treated L5178YR tumor cells (PKHB1-TCL) was able to induce, ex vivo, dendritic cells maturation, cytokine production, and T cell antitumor responses. Finally, our results show that in vivo, PKHB1-TCL treatment induces tumor regression in syngeneic mice transplanted with L5178Y-R cells, increasing their overall survival and protecting them from further tumor establishment after tumor rechallenge. Altogether our results highlight the immunogenicity of the cell death induced by PKHB1 activation of CD47 as a potential therapeutic tool to overcome the low immunogenicity and immune tolerance in T-ALL.


Sign in / Sign up

Export Citation Format

Share Document